1
|
Kent A, Yee Mon KJ, Hutchins Z, Putzel G, Zhigarev D, Grier A, Jia B, Kortlever RM, Barbet G, Evan GI, Blander JM. A stromal inflammasome Ras safeguard against Myc-driven lymphomagenesis. Nat Immunol 2025; 26:53-67. [PMID: 39747433 DOI: 10.1038/s41590-024-02028-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 10/29/2024] [Indexed: 01/04/2025]
Abstract
The inflammasome plays multifaceted roles in cancer, but less is known about its function during premalignancy upon initial cell transformation. We report a homeostatic function of the inflammasome in suppressing malignant transformation through Ras inhibition. We identified increased hematopoietic stem cell (HSC) proliferation within the bone marrow of inflammasome-deficient mice. HSCs within an inflammasome-deficient stroma expressed a Ras signature associated with increased Ras pathway- and cancer-related transcripts and heightened levels of cytokine, chemokine and growth factor receptors. Stromal inflammasome deficiency established a poised Ras-dependent mitogenic state within HSCs, which fueled progeny B cell lymphomagenesis upon Myc deregulation in a spontaneous model of B cell lymphoma, and shortened its premalignant stage leading to faster onset of malignancy. Thus, the stromal inflammasome preserves tissue balance by restraining Ras to disrupt the most common oncogenic Myc-Ras cooperation and establish a natural defense against transition to malignancy. These findings should inform preventative therapies against hematological malignancies.
Collapse
Affiliation(s)
- Andrew Kent
- Division of Hematology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kristel Joy Yee Mon
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Zachary Hutchins
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Immunology and Microbial Pathogenesis, College of Life Sciences, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Gregory Putzel
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Microbial Computational Genomic Core Lab, Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Dmitry Zhigarev
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Wistar Institute, Philadelphia, PA, USA
| | - Alexander Grier
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Baosen Jia
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Gaetan Barbet
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Pediatrics, The Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Gerard I Evan
- The Francis Crick Institute, London, UK
- Kings College London, London, UK
| | - J Magarian Blander
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Immunology and Microbial Pathogenesis Program, Weill Cornell and Sloan Kettering Institute Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
2
|
Sharifi MJ, Xu L, Nasiri N, Ashja‐Arvan M, Soleimanzadeh H, Ganjalikhani‐Hakemi M. Immune-dysregulation harnessing in myeloid neoplasms. Cancer Med 2024; 13:e70152. [PMID: 39254117 PMCID: PMC11386321 DOI: 10.1002/cam4.70152] [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: 05/05/2024] [Revised: 08/02/2024] [Accepted: 08/16/2024] [Indexed: 09/11/2024] Open
Abstract
Myeloid malignancies arise in bone marrow microenvironments and shape these microenvironments in favor of malignant development. Immune suppression is one of the most important stages in myeloid leukemia progression. Leukemic clone expansion and immune dysregulation occur simultaneously in bone marrow microenvironments. Complex interactions emerge between normal immune system elements and leukemic clones in the bone marrow. In recent years, researchers have identified several of these pathological interactions. For instance, recent works shows that the secretion of inflammatory cytokines such as tumor necrosis factor-α (TNF-α), from bone marrow stromal cells contributes to immune dysregulation and the selective proliferation of JAK2V617F+ clones in myeloproliferative neoplasms. Moreover, inflammasome activation and sterile inflammation result in inflamed microenvironments and the development of myelodysplastic syndromes. Additional immune dysregulations, such as exhaustion of T and NK cells, an increase in regulatory T cells, and impairments in antigen presentation are common findings in myeloid malignancies. In this review, we discuss the role of altered bone marrow microenvironments in the induction of immune dysregulations that accompany myeloid malignancies. We also consider both current and novel therapeutic strategies to restore normal immune system function in the context of myeloid malignancies.
Collapse
Affiliation(s)
- Mohammad Jafar Sharifi
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences, School of Paramedical SciencesShiraz University of Medical SciencesShirazIran
| | - Ling Xu
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan UniversityGuangzhouChina
| | - Nahid Nasiri
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences, School of Paramedical SciencesShiraz University of Medical SciencesShirazIran
| | - Mehnoosh Ashja‐Arvan
- Regenerative and Restorative Medicine Research Center (REMER)Research Institute of Health sciences and Technology (SABITA), Istanbul Medipol UniversityIstanbulTurkey
| | - Hadis Soleimanzadeh
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences, School of Paramedical SciencesShiraz University of Medical SciencesShirazIran
| | - Mazdak Ganjalikhani‐Hakemi
- Regenerative and Restorative Medicine Research Center (REMER)Research Institute of Health sciences and Technology (SABITA), Istanbul Medipol UniversityIstanbulTurkey
- Department of Immunology, Faculty of MedicineIsfahan University of Medical SciencesIsfahanIran
| |
Collapse
|
3
|
Khattab S, El Sorady M, El-Ghandour A, Visani G, Piccaluga PP. Hematopoietic and leukemic stem cells homeostasis: the role of bone marrow niche. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:1027-1055. [PMID: 39351440 PMCID: PMC11438561 DOI: 10.37349/etat.2024.00262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 07/01/2024] [Indexed: 10/04/2024] Open
Abstract
The bone marrow microenvironment (BMM) has highly specialized anatomical characteristics that provide a sanctuary place for hematopoietic stem cells (HSCs) that allow appropriate proliferation, maintenance, and self-renewal capacity. Several cell types contribute to the constitution and function of the bone marrow niche. Interestingly, uncovering the secrets of BMM and its interaction with HSCs in health paved the road for research aiming at better understanding the concept of leukemic stem cells (LSCs) and their altered niche. In fact, they share many signals that are responsible for interactions between LSCs and the bone marrow niche, due to several biological similarities between LSCs and HSCs. On the other hand, LSCs differ from HSCs in their abnormal activation of important signaling pathways that regulate survival, proliferation, drug resistance, invasion, and spread. Targeting these altered niches can help in better treatment choices for hematological malignancies and bone marrow disorders in general and acute myeloid leukemia (AML) in particular. Moreover, targeting those niches may help in decreasing the emergence of drug resistance and lower the relapse rate. In this article, the authors reviewed the most recent literature on bone marrow niches and their relations with either normal HSCs and AML cells/LSC, by focusing on pathogenetic and therapeutic implications.
Collapse
Affiliation(s)
- Shaimaa Khattab
- Biobank of Research, IRCCS Azienda Ospedaliera-Universitaria di Bologna Policlinico di S. Orsola, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences, Bologna University School of Medicine, 40138 Bologna, Italy
- Medical Research Institute, Hematology department, Alexandria University, Alexandria 21561, Egypt
| | - Manal El Sorady
- Department of Internal Medicine, Faculty of Medicine, Alexandria University, Alexandria 5310002, Egypt
| | - Ashraf El-Ghandour
- Department of Internal Medicine, Faculty of Medicine, Alexandria University, Alexandria 5310002, Egypt
| | - Giuseppe Visani
- Hematology and Stem Cell Transplant Center, Azienda Ospedaliera Marche Nord, 61121 Pesaro, Italy
| | - Pier Paolo Piccaluga
- Biobank of Research, IRCCS Azienda Ospedaliera-Universitaria di Bologna Policlinico di S. Orsola, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences, Bologna University School of Medicine, 40138 Bologna, Italy
| |
Collapse
|
4
|
Li Z, Yao X, Zhang J, Yang J, Ni J, Wang Y. Exploring the bone marrow micro environment in thalassemia patients: potential therapeutic alternatives. Front Immunol 2024; 15:1403458. [PMID: 39161767 PMCID: PMC11330836 DOI: 10.3389/fimmu.2024.1403458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 07/22/2024] [Indexed: 08/21/2024] Open
Abstract
Genetic mutations in the β-globin gene lead to a decrease or removal of the β-globin chain, causing the build-up of unstable alpha-hemoglobin. This condition is referred to as beta-thalassemia (BT). The present treatment strategies primarily target the correction of defective erythropoiesis, with a particular emphasis on gene therapy and hematopoietic stem cell transplantation. However, the presence of inefficient erythropoiesis in BT bone marrow (BM) is likely to disturb the previously functioning BM microenvironment. This includes accumulation of various macromolecules, damage to hematopoietic function, destruction of bone cell production and damage to osteoblast(OBs), and so on. In addition, the changes of BT BM microenvironment may have a certain correlation with the occurrence of hematological malignancies. Correction of the microenvironment can be achieved through treatments such as iron chelation, antioxidants, hypoglycemia, and biologics. Hence, This review describes damage in the BT BM microenvironment and some potential remedies.
Collapse
Affiliation(s)
- Zengzheng Li
- Department of Hematology, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Province Clinical Research Center for Hematologic Disease, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
- Yunnan Provincial Clinical Medical Center for Blood Diseases and Thrombosis Prevention and Treatment, Kunming, Yunnan, China
| | - Xiangmei Yao
- Department of Hematology, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Province Clinical Research Center for Hematologic Disease, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
- Yunnan Provincial Clinical Medical Center for Blood Diseases and Thrombosis Prevention and Treatment, Kunming, Yunnan, China
| | - Jie Zhang
- Department of Medical Genetics, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jinghui Yang
- Department of Pediatrics, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Junxue Ni
- Hospital Office, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Yajie Wang
- Department of Hematology, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Province Clinical Research Center for Hematologic Disease, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
- Yunnan Provincial Clinical Medical Center for Blood Diseases and Thrombosis Prevention and Treatment, Kunming, Yunnan, China
| |
Collapse
|
5
|
Wei D, Liang X, Huang M, Wang C, Ye Z, Zhang T, Zhang J. Targeting histone deacetylase 1 (HDAC1) in the bone marrow stromal cells revers imatinib resistance by modulating IL-6 in Ph + acute lymphoblastic leukemia. Ann Hematol 2024; 103:3015-3027. [PMID: 38847852 DOI: 10.1007/s00277-024-05830-9] [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: 11/27/2023] [Accepted: 05/31/2024] [Indexed: 07/28/2024]
Abstract
Bone marrow stromal cells (BMSCs) can promote the growth of Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). Histone deacetylases (HDACs) play essential roles in the proliferation and apoptosis resistance of Ph + ALL cells. In our previous study, inhibiting histone deacetylase 1 (HDAC1) decreases the proliferation of Ph + ALL cells. However, little is known regarding how HDAC1 in BMSCs of Ph + ALL patients affects the imatinib (IM) resistance. Therefore, the present work examined the roles of HDAC1 in BMSCs. Overexpression of HDAC1 was found in BMSCs of Ph + ALL patients with IM resistance. In addition, the Ph + ALL cell line SUP-B15 was co-cultured with BMSCs after lentivirus transfection for regulating HDAC1 expression. Knockdown of HDAC1 within BMSCs elevated the IM-mediated SUP-B15 cell apoptosis, while increasing HDAC1 expression had an opposite effect. IL-6 in BMSCs, which is an important factor for the microenvironment-associated chemoresistance, showed evident up-regulation in HDAC1-upregulated BMSCs and down-regulation in HDAC1-downregulated BMSCs. While recombinant IL-6 (rIL-6) can reversed the sensitivity of SUP-B15 cells to IM induced by downregulating HDAC1 expression in BMSCs. HDAC1 showed positive regulation on IL-6 transcription and secretion. Moreover, IL-6 secretion induced by HDAC1 in BMSCs might enhance IM resistance in Ph + ALL cells. With regard to the underlying molecular mechanism, NF-κB, an important signal responsible for IL-6 transcription in BMSCs, mediated the HDAC1-regulated IL-6 expression. Collectively, this study facilitated to develop HDAC1 inhibitors based not only the corresponding direct anti-Ph + ALL activity but also the regulation of bone marrow microenvironment.
Collapse
Affiliation(s)
- Danna Wei
- Department of Pediatric Hematology, Guiyang Maternal and Child Health Care Hospital, Guiyang Children's Hospital, Guiyang, 550002, China
| | - Xiaoling Liang
- Department of Pediatric Hematology, Guiyang Maternal and Child Health Care Hospital, Guiyang Children's Hospital, Guiyang, 550002, China
| | - Meiling Huang
- Department of Pediatric Hematology, Guiyang Maternal and Child Health Care Hospital, Guiyang Children's Hospital, Guiyang, 550002, China
| | - Caili Wang
- Department of Pediatric Hematology, Guiyang Maternal and Child Health Care Hospital, Guiyang Children's Hospital, Guiyang, 550002, China
| | - Zhangmin Ye
- Department of Pediatric Hematology, Guiyang Maternal and Child Health Care Hospital, Guiyang Children's Hospital, Guiyang, 550002, China
| | - Tianzhuo Zhang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
| | - Jingrong Zhang
- Department of Pediatric Hematology, Guiyang Maternal and Child Health Care Hospital, Guiyang Children's Hospital, Guiyang, 550002, China.
| |
Collapse
|
6
|
Lana JF, Navani A, Jeyaraman M, Santos N, Pires L, Santos GS, Rodrigues IJ, Santos D, Mosaner T, Azzini G, da Fonseca LF, de Macedo AP, Huber SC, de Moraes Ferreira Jorge D, Purita J. Sacral Bioneuromodulation: The Role of Bone Marrow Aspirate in Spinal Cord Injuries. Bioengineering (Basel) 2024; 11:461. [PMID: 38790327 PMCID: PMC11118755 DOI: 10.3390/bioengineering11050461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
Spinal cord injury (SCI) represents a severe trauma to the nervous system, leading to significant neurological damage, chronic inflammation, and persistent neuropathic pain. Current treatments, including pharmacotherapy, immobilization, physical therapy, and surgical interventions, often fall short in fully addressing the underlying pathophysiology and resultant disabilities. Emerging research in the field of regenerative medicine has introduced innovative approaches such as autologous orthobiologic therapies, with bone marrow aspirate (BMA) being particularly notable for its regenerative and anti-inflammatory properties. This review focuses on the potential of BMA to modulate inflammatory pathways, enhance tissue regeneration, and restore neurological function disrupted by SCI. We hypothesize that BMA's bioactive components may stimulate reparative processes at the cellular level, particularly when applied at strategic sites like the sacral hiatus to influence lumbar centers and higher neurological structures. By exploring the mechanisms through which BMA influences spinal repair, this review aims to establish a foundation for its application in clinical settings, potentially offering a transformative approach to SCI management that extends beyond symptomatic relief to promoting functional recovery.
Collapse
Affiliation(s)
- José Fábio Lana
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
- Clinical Research, Anna Vitória Lana Institute (IAVL), Indaiatuba 13334-170, SP, Brazil
- Medical School, Max Planck University Center (UniMAX), Indaiatuba 13343-060, SP, Brazil
| | - Annu Navani
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
- Medical School, Max Planck University Center (UniMAX), Indaiatuba 13343-060, SP, Brazil
- Comprehensive Spine & Sports Center, Campbell, CA 95008, USA
| | - Madhan Jeyaraman
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
- Department of Orthopaedics, ACS Medical College and Hospital, Chennai 600077, Tamil Nadu, India
| | - Napoliane Santos
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Luyddy Pires
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Gabriel Silva Santos
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Izair Jefthé Rodrigues
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Douglas Santos
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Tomas Mosaner
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Gabriel Azzini
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Lucas Furtado da Fonseca
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
- Medical School, Federal University of São Paulo (UNIFESP), São Paulo 04024-002, SP, Brazil
| | - Alex Pontes de Macedo
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Stephany Cares Huber
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Daniel de Moraes Ferreira Jorge
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Joseph Purita
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
- Medical School, Max Planck University Center (UniMAX), Indaiatuba 13343-060, SP, Brazil
| |
Collapse
|
7
|
Sharma NS, Choudhary B. Good Cop, Bad Cop: Profiling the Immune Landscape in Multiple Myeloma. Biomolecules 2023; 13:1629. [PMID: 38002311 PMCID: PMC10669790 DOI: 10.3390/biom13111629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/26/2023] [Accepted: 10/29/2023] [Indexed: 11/26/2023] Open
Abstract
Multiple myeloma (MM) is a dyscrasia of plasma cells (PCs) characterized by abnormal immunoglobulin (Ig) production. The disease remains incurable due to a multitude of mutations and structural abnormalities in MM cells, coupled with a favorable microenvironment and immune suppression that eventually contribute to the development of drug resistance. The bone marrow microenvironment (BMME) is composed of a cellular component comprising stromal cells, endothelial cells, osteoclasts, osteoblasts, and immune cells, and a non-cellular component made of the extracellular matrix (ECM) and the liquid milieu, which contains cytokines, growth factors, and chemokines. The bone marrow stromal cells (BMSCs) are involved in the adhesion of MM cells, promote the growth, proliferation, invasion, and drug resistance of MM cells, and are also crucial in angiogenesis and the formation of lytic bone lesions. Classical immunophenotyping in combination with advanced immune profiling using single-cell sequencing technologies has enabled immune cell-specific gene expression analysis in MM to further elucidate the roles of specific immune cell fractions from peripheral blood and bone marrow (BM) in myelomagenesis and progression, immune evasion and exhaustion mechanisms, and development of drug resistance and relapse. The review describes the role of BMME components in MM development and ongoing clinical trials using immunotherapeutic approaches.
Collapse
Affiliation(s)
- Niyati Seshagiri Sharma
- Institute of Bioinformatics and Applied Biotechnology (IBAB), Electronic City, Bengaluru 560100, India
- Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | - Bibha Choudhary
- Institute of Bioinformatics and Applied Biotechnology (IBAB), Electronic City, Bengaluru 560100, India
| |
Collapse
|
8
|
Pottosin I, Olivas-Aguirre M, Dobrovinskaya O. In vitro simulation of the acute lymphoblastic leukemia niche: a critical view on the optimal approximation for drug testing. J Leukoc Biol 2023; 114:21-41. [PMID: 37039524 DOI: 10.1093/jleuko/qiad039] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 03/29/2023] [Accepted: 03/31/2023] [Indexed: 04/12/2023] Open
Abstract
Acute lymphoblastic leukemia with the worst prognosis is related to minimal residual disease. Minimal residual disease not only depends on the individual peculiarities of leukemic clones but also reflects the protective role of the acute lymphoblastic leukemia microenvironment. In this review, we discuss in detail cell-to-cell interactions in the 2 leukemic niches, more explored bone marrow and less studied extramedullary adipose tissue. A special emphasis is given to multiple ways of interactions of acute lymphoblastic leukemia cells with the bone marrow or extramedullary adipose tissue microenvironment, indicating observed differences in B- and T-cell-derived acute lymphoblastic leukemia behavior. This analysis argued for the usage of coculture systems for drug testing. Starting with a review of available sources and characteristics of acute lymphoblastic leukemia cells, mesenchymal stromal cells, endothelial cells, and adipocytes, we have then made an update of the available 2-dimensional and 3-dimensional systems, which bring together cellular elements, components of the extracellular matrix, or its imitation. We discussed the most complex available 3-dimensional systems like "leukemia-on-a-chip," which include either a prefabricated microfluidics platform or, alternatively, the microarchitecture, designed by using the 3-dimensional bioprinting technologies. From our analysis, it follows that for preclinical antileukemic drug testing, in most cases, intermediately complex in vitro cell systems are optimal, such as a "2.5-dimensional" coculture of acute lymphoblastic leukemia cells with niche cells (mesenchymal stromal cells, endothelial cells) plus matrix components or scaffold-free mesenchymal stromal cell organoids, populated by acute lymphoblastic leukemia cells. Due to emerging evidence for the correlation of obesity and poor prognosis, a coculture of adipocytes with acute lymphoblastic leukemia cells as a drug testing system is gaining shape.
Collapse
Affiliation(s)
- Igor Pottosin
- Laboratory of Immunobiology and Ionic Transport Regulation, University Center for Biomedical Research, University of Colima, Av. Enrique Arreola Silva 883, Guzmán City, Jalisco, 49000, Mexico
| | - Miguel Olivas-Aguirre
- Laboratory of Immunobiology and Ionic Transport Regulation, University Center for Biomedical Research, University of Colima, Av. Enrique Arreola Silva 883, Guzmán City, Jalisco, 49000, Mexico
- Division of Exact, Natural and Technological Sciences, South University Center (CUSUR), University of Guadalajara, Jalisco, Mexico
| | - Oxana Dobrovinskaya
- Laboratory of Immunobiology and Ionic Transport Regulation, University Center for Biomedical Research, University of Colima, Av. Enrique Arreola Silva 883, Guzmán City, Jalisco, 49000, Mexico
| |
Collapse
|
9
|
Romano IR, D'Angeli F, Vicario N, Russo C, Genovese C, Lo Furno D, Mannino G, Tamburino S, Parenti R, Giuffrida R. Adipose-Derived Mesenchymal Stromal Cells: A Tool for Bone and Cartilage Repair. Biomedicines 2023; 11:1781. [PMID: 37509421 PMCID: PMC10376676 DOI: 10.3390/biomedicines11071781] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
The osteogenic and chondrogenic differentiation ability of adipose-derived mesenchymal stromal cells (ASCs) and their potential therapeutic applications in bone and cartilage defects are reported in this review. This becomes particularly important when these disorders can only be poorly treated by conventional therapeutic approaches, and tissue engineering may represent a valuable alternative. Being of mesodermal origin, ASCs can be easily induced to differentiate into chondrocyte-like and osteocyte-like elements and used to repair damaged tissues. Moreover, they can be easily harvested and used for autologous implantation. A plethora of ASC-based strategies are being developed worldwide: they include the transplantation of freshly harvested cells, in vitro expanded cells or predifferentiated cells. Moreover, improving their positive effects, ASCs can be implanted in combination with several types of scaffolds that ensure the correct cell positioning; support cell viability, proliferation and migration; and may contribute to their osteogenic or chondrogenic differentiation. Examples of these strategies are described here, showing the enormous therapeutic potential of ASCs in this field. For safety and regulatory issues, most investigations are still at the experimental stage and carried out in vitro and in animal models. Clinical applications have, however, been reported with promising results and no serious adverse effects.
Collapse
Affiliation(s)
- Ivana Roberta Romano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Floriana D'Angeli
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Roma Open University, 00166 Rome, Italy
| | - Nunzio Vicario
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Cristina Russo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Carlo Genovese
- Faculty of Medicine and Surgery, "Kore" University of Enna, 94100 Enna, Italy
| | - Debora Lo Furno
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Giuliana Mannino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98122 Messina, Italy
| | - Serena Tamburino
- Chi.Pla Chirurgia Plastica, Via Suor Maria Mazzarello, 54, 95128 Catania, Italy
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Rosario Giuffrida
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| |
Collapse
|
10
|
Wu Y, Shi X, Yao X, Du X. Biological research on the occurrence and development of multiple myeloma and its treatment. Immun Inflamm Dis 2023; 11:e850. [PMID: 37249283 PMCID: PMC10165958 DOI: 10.1002/iid3.850] [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: 02/06/2023] [Revised: 03/30/2023] [Accepted: 04/16/2023] [Indexed: 05/31/2023] Open
Abstract
INTRODUCTION To review the pathogenesis and treatment of multiple myeloma (MM). MM is a hematological malignancy with abnormal plasma cell proliferation in bone marrow. Due to the emergence of drug resistance, MM is still an incurable malignancy, which requires further exploration of pathogenesis and effective therapeutic targets. METHODS In this paper, the method of literature review is adopted to obtain the information about MM. Based on the literature, comprehensive and systematic review is made. RESULTS MM is a complex pathophysiological process with great heterogeneity, mainly reflected in genomic instability and bone marrow microenvironment. At present, the treatment of MM has made great progress, proteasome inhibitors and immunomodulatory drugs are widely used in clinic. Allogeneic stem cell transplantation may be the only promising cure for MM, and its high transplant-related mortality limits its clinical application. CONCLUSIONS The future of MM treatment lies in the development of more targeted therapies, novel immunotherapies, and a better understanding of the disease's molecular and genetic basis.
Collapse
Affiliation(s)
- Yue Wu
- Department of OrthopaedicsBeijing Chao‐Yang HospitalBeijingChina
| | - Xiangjun Shi
- Department of OrthopaedicsBeijing Chao‐Yang HospitalBeijingChina
| | - Xinchen Yao
- Department of OrthopaedicsBeijing Chao‐Yang HospitalBeijingChina
| | - Xinru Du
- Department of OrthopaedicsBeijing Chao‐Yang HospitalBeijingChina
| |
Collapse
|
11
|
Dausinas Ni P, Hartman M, Slack J, Basile C, Liu S, Wan J, O'Leary HA. Novel differential calcium regulation of hematopoietic stem and progenitor cells under physiological low oxygen conditions. J Cell Physiol 2023. [PMID: 37051890 DOI: 10.1002/jcp.30942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 11/28/2022] [Accepted: 12/23/2022] [Indexed: 04/14/2023]
Abstract
Low oxygen bone marrow (BM) niches (~1%-4% low O2 ) provide critical signals for hematopoietic stem/progenitor cells (HSC/HSPCs). Our presented data are the first to investigate live, sorted HSC/HSPCs in their native low O2 conditions. Transcriptional and proteomic analysis uncovered differential Ca2+ regulation that correlated with overlapping phenotypic populations consisting of robust increases of cytosolic and mitochondrial Ca2+ , ABC transporter (ABCG2) expression and sodium/hydrogen exchanger (NHE1) expression in live, HSC/HSPCs remaining in constant low O2. We identified a novel Ca2+ high population in HSPCs predominantly detected in low O2 that displayed enhanced frequency of phenotypic LSK/LSKCD150 in low O2 replating assays compared to Ca2+ low populations. Inhibition of the Ca2+ regulator NHE1 (Cariporide) resulted in attenuation of both the low O2 induced Ca2+ high population and subsequent enhanced maintenance of phenotypic LSK and LSKCD150 during low O2 replating. These data reveal multiple levels of differential Ca2+ regulation in low O2 resulting in phenotypic, signaling, and functional consequences in HSC/HSPCs.
Collapse
Affiliation(s)
- Paige Dausinas Ni
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Melissa Hartman
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jacob Slack
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Christopher Basile
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Center of Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Center of Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Heather A O'Leary
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| |
Collapse
|
12
|
Zappalà A, Romano IR, D’Angeli F, Musumeci G, Lo Furno D, Giuffrida R, Mannino G. Functional Roles of Connexins and Gap Junctions in Osteo-Chondral Cellular Components. Int J Mol Sci 2023; 24:ijms24044156. [PMID: 36835567 PMCID: PMC9967557 DOI: 10.3390/ijms24044156] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Gap junctions (GJs) formed by connexins (Cxs) play an important role in the intercellular communication within most body tissues. In this paper, we focus on GJs and Cxs present in skeletal tissues. Cx43 is the most expressed connexin, participating in the formation of both GJs for intercellular communication and hemichannels (HCs) for communication with the external environment. Through GJs in long dendritic-like cytoplasmic processes, osteocytes embedded in deep lacunae are able to form a functional syncytium not only with neighboring osteocytes but also with bone cells located at the bone surface, despite the surrounding mineralized matrix. The functional syncytium allows a coordinated cell activity through the wide propagation of calcium waves, nutrients and anabolic and/or catabolic factors. Acting as mechanosensors, osteocytes are able to transduce mechanical stimuli into biological signals that spread through the syncytium to orchestrate bone remodeling. The fundamental role of Cxs and GJs is confirmed by a plethora of investigations that have highlighted how up- and downregulation of Cxs and GJs critically influence skeletal development and cartilage functions. A better knowledge of GJ and Cx mechanisms in physiological and pathological conditions might help in developing therapeutic approaches aimed at the treatment of human skeletal system disorders.
Collapse
Affiliation(s)
- Agata Zappalà
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Ivana Roberta Romano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Floriana D’Angeli
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Roma Open University, 00166 Rome, Italy
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Debora Lo Furno
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
- Correspondence: (D.L.F.); (R.G.)
| | - Rosario Giuffrida
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
- Correspondence: (D.L.F.); (R.G.)
| | - Giuliana Mannino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98122 Messina, Italy
| |
Collapse
|
13
|
van Santen VJB, Zandieh Doulabi B, Semeins CM, Hogervorst JMA, Bratengeier C, Bakker AD. Compressed Prostate Cancer Cells Decrease Osteoclast Activity While Enhancing Osteoblast Activity In Vitro. Int J Mol Sci 2023; 24:ijms24010759. [PMID: 36614201 PMCID: PMC9821660 DOI: 10.3390/ijms24010759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/21/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023] Open
Abstract
Once prostate cancer cells metastasize to bone, they perceive approximately 2 kPa compression. We hypothesize that 2 kPa compression stimulates the epithelial-to-mesenchymal transition (EMT) of prostate cancer cells and alters their production of paracrine signals to affect osteoclast and osteoblast behavior. Human DU145 prostate cancer cells were subjected to 2 kPa compression for 2 days. Compression decreased expression of 2 epithelial genes, 5 out of 13 mesenchymal genes, and increased 2 mesenchymal genes by DU145 cells, as quantified by qPCR. Conditioned medium (CM) of DU145 cells was added to human monocytes that were stimulated to differentiate into osteoclasts for 21 days. CM from compressed DU145 cells decreased osteoclast resorptive activity by 38% but did not affect osteoclast size and number compared to CM from non-compressed cells. CM was also added to human adipose stromal cells, grown in osteogenic medium. CM of compressed DU145 cells increased bone nodule production (Alizarin Red) by osteoblasts from four out of six donors. Compression did not affect IL6 or TNF-α production by PC DU145 cells. Our data suggest that compression affects EMT-related gene expression in DU145 cells, and alters their production of paracrine signals to decrease osteoclast resorptive activity while increasing mineralization by osteoblasts is donor dependent. This observation gives further insight in the altered behavior of PC cells upon mechanical stimuli, which could provide novel leads for therapies, preventing bone metastases.
Collapse
Affiliation(s)
- Victor J. B. van Santen
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, 1081 LA Amsterdam, The Netherlands
| | - Behrouz Zandieh Doulabi
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, 1081 LA Amsterdam, The Netherlands
| | - Cornelis M. Semeins
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, 1081 LA Amsterdam, The Netherlands
| | - Jolanda M. A. Hogervorst
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, 1081 LA Amsterdam, The Netherlands
| | - Cornelia Bratengeier
- Division of Cell Biology, Department of Clinical and Experimental Medicine, Linköping University, SE-581 83 Linköping, Sweden
| | - Astrid D. Bakker
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, 1081 LA Amsterdam, The Netherlands
- Correspondence: ; Tel.: +31-(0)20-5980224
| |
Collapse
|
14
|
Zhang H, Linster E, Wirtz M, Theodoulou FL. Relative Protein Lifetime Measurement in Plants Using Tandem Fluorescent Protein Timers. Methods Mol Biol 2023; 2581:201-220. [PMID: 36413319 DOI: 10.1007/978-1-0716-2784-6_14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Targeted protein degradation plays a wide range of important roles in plant growth and development, but analyzing protein turnover in vivo is technically challenging. Until recently, there has been no straightforward methodology for quantifying protein dynamics at subcellular resolution during cellular transitions in plants. A tandem fluorescent protein timer (tFT) is a fusion of two different fluorescent proteins with distinct fluorophore maturation kinetics, which allows estimation of relative protein age from the ratio of fluorescence intensities of the two fluorescent proteins. Here, we describe approaches to use this technology to report relative protein lifetime in both transient and stable plant transformation systems. tFTs enable in vivo, real-time protein lifetime assessment within subcellular compartments and across tissues, permitting the analysis of protein degradation dynamics in response to stresses or developmental cues and in different genetic backgrounds.
Collapse
Affiliation(s)
- Hongtao Zhang
- Plant Sciences and the Bioeconomy, Rothamsted Research, Harpenden, UK
| | - Eric Linster
- Centre for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Markus Wirtz
- Centre for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | | |
Collapse
|
15
|
Brum da Silva Nunes V, Kehl Dias C, Nathali Scholl J, Nedel Sant'Ana A, de Fraga Dias A, Granero Farias M, Alegretti AP, Sosnoski M, Esteves Daudt L, Bohns Michalowski M, Oliveira Battastini AM, Paz AA, Figueiró F. Lymphocytes from B-acute lymphoblastic leukemia patients present differential regulation of the adenosinergic axis depending on risk stratification. Discov Oncol 2022; 13:143. [PMID: 36581667 PMCID: PMC9800668 DOI: 10.1007/s12672-022-00602-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/08/2022] [Indexed: 12/31/2022] Open
Abstract
PURPOSE Although risk-stratified chemotherapy regimens improve B-cell acute lymphoblastic leukemia (B-ALL) clinical outcome, relapse occurs in a significant number of cases. The identification of new therapeutic targets as well as prognostic and diagnostic biomarkers can improve B-ALL patients' clinical outcomes. Purinergic signaling is an important pathway in cancer progression, however the expression of ectonucleotidases and their impact on immune cells in B-ALL lacks exploration. We aimed to analyze the expression of ectonucleotidases in B-ALL patients' lymphocyte subpopulations. METHODS Peripheral blood samples from 15 patients diagnosed with B-ALL were analyzed. Flow cytometry was used to analyze cellularity, expression level of CD38, CD39, and CD73, and frequency of [Formula: see text], and [Formula: see text] in lymphocyte subpopulations. Plasma was used for cytokines (by CBA kit) and adenine nucleosides/nucleotides detection (by HPLC). RESULTS Comparing B-ALL patients to health donors, we observed an increase of CD4 + and CD8 + T-cells. In addition, a decrease in CD38 expression in B and Treg subpopulations and an increase in CD39+ CD73+ frequency in Breg and CD8+ T-cells. Analyzing cytokines and adenine nucleosides/nucleotides, we found a decrease in TNF, IL-1β, and ADO concentrations, together with an increase in AMP in B-ALL patients' plasma. CONCLUSION As immunomodulators, the expression of ectonucleotidases might be associated with activation states, as well as the abundance of different cellular subsets. We observed a pro-tumor immunity expression profile in B-ALL patients at diagnosis, being associated with cell exhaustion and immune evasion in B-ALL.
Collapse
Affiliation(s)
- Vitória Brum da Silva Nunes
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | - Camila Kehl Dias
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | - Juliete Nathali Scholl
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | - Alexia Nedel Sant'Ana
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | - Amanda de Fraga Dias
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | | | - Ana Paula Alegretti
- Hospital de Clínicas de Porto Alegre/HCPA, Porto Alegre, RS, CEP 90035-903, Brazil
| | - Monalisa Sosnoski
- Hospital de Clínicas de Porto Alegre/HCPA, Porto Alegre, RS, CEP 90035-903, Brazil
| | - Liane Esteves Daudt
- Hospital de Clínicas de Porto Alegre/HCPA, Porto Alegre, RS, CEP 90035-903, Brazil
| | - Mariana Bohns Michalowski
- Hospital de Clínicas de Porto Alegre/HCPA, Porto Alegre, RS, CEP 90035-903, Brazil
- Programa de Pós-Graduação em Saúde da Criança e do Adolescente, Faculdade de Medicina, UFRGS, Porto Alegre, RS, 90035-003, Brazil
| | - Ana Maria Oliveira Battastini
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | | | - Fabrício Figueiró
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil.
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil.
| |
Collapse
|
16
|
Molecular Features of the Mesenchymal and Osteoblastic Cells in Multiple Myeloma. Int J Mol Sci 2022; 23:ijms232415448. [PMID: 36555090 PMCID: PMC9779562 DOI: 10.3390/ijms232415448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/02/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma (MM) is a monoclonal gammopathy characterized by biological heterogeneity and unregulated proliferation of plasma cells (PCs) in bone marrow (BM). MM is a multistep process based on genomic instability, epigenetic dysregulation and a tight cross-talk with the BM microenvironment that plays a pivotal role supporting the proliferation, survival, drug-resistance and homing of PCs. The BM microenvironment consists of a hematopoietic and a non-hematopoietic compartment, which cooperate to create a tumor environment. Among the non-hematopoietic component, mesenchymal stromal cells (MSCs) and osteoblasts (OBs) appear transcriptionally and functionally different in MM patients compared to healthy donors (HDs) and to patients with pre-malignant monoclonal gammopathies. Alterations of both MSCs and OBs underly the osteolytic lesions that characterize myeloma-associated bone disease. In this review, we will discuss the different characteristics of MSCs and OBs in MM patients, analyzing the transcriptome, the deregulated molecular pathways and the role performed by miRNAs and exosome in the pathophysiology of MM.
Collapse
|
17
|
Zhang Z, Yang K, Zhang H. Targeting Leukemia-Initiating Cells and Leukemic Niches: The Next Therapy Station for T-Cell Acute Lymphoblastic Leukemia? Cancers (Basel) 2022; 14:cancers14225655. [PMID: 36428753 PMCID: PMC9688677 DOI: 10.3390/cancers14225655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/09/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive subtype of hematological malignancy characterized by its high heterogeneity and potentially life-threatening clinical features. Despite the advances in risk stratification and therapeutic management of T-ALL, patients often suffer from treatment failure and chemotherapy-induced toxicity, calling for greater efforts to improve therapeutic efficacy and safety in the treatment of T-ALL. During the past decades, increasing evidence has shown the indispensable effects of leukemia-initiating cells (LICs) and leukemic niches on T-ALL initiation and progression. These milestones greatly facilitate precision medicine by interfering with the pathways that are associated with LICs and leukemic niches or by targeting themselves directly. Most of these novel agents, either alone or in combination with conventional chemotherapy, have shown promising preclinical results, facilitating them to be further evaluated under clinical trials. In this review, we summarize the latest discoveries in LICs and leukemic niches in terms of T-ALL, with a particular highlight on the current precision medicine. The challenges and future prospects are also discussed.
Collapse
Affiliation(s)
- Ziting Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Kun Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
- School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Han Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
- Correspondence: ; Tel.: +86-158-7796-3252
| |
Collapse
|
18
|
Lourenço D, Lopes R, Pestana C, Queirós AC, João C, Carneiro EA. Patient-Derived Multiple Myeloma 3D Models for Personalized Medicine-Are We There Yet? Int J Mol Sci 2022; 23:12888. [PMID: 36361677 PMCID: PMC9657251 DOI: 10.3390/ijms232112888] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/19/2022] [Accepted: 10/22/2022] [Indexed: 12/03/2023] Open
Abstract
Despite the wide variety of existing therapies, multiple myeloma (MM) remains a disease with dismal prognosis. Choosing the right treatment for each patient remains one of the major challenges. A new approach being explored is the use of ex vivo models for personalized medicine. Two-dimensional culture or animal models often fail to predict clinical outcomes. Three-dimensional ex vivo models using patients' bone marrow (BM) cells may better reproduce the complexity and heterogeneity of the BM microenvironment. Here, we review the strengths and limitations of currently existing patient-derived ex vivo three-dimensional MM models. We analyze their biochemical and biophysical properties, molecular and cellular characteristics, as well as their potential for drug testing and identification of disease biomarkers. Furthermore, we discuss the remaining challenges and give some insight on how to achieve a more biomimetic and accurate MM BM model. Overall, there is still a need for standardized culture methods and refined readout techniques. Including both myeloma and other cells of the BM microenvironment in a simple and reproducible three-dimensional scaffold is the key to faithfully mapping and examining the relationship between these players in MM. This will allow a patient-personalized profile, providing a powerful tool for clinical and research applications.
Collapse
Affiliation(s)
- Diana Lourenço
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Raquel Lopes
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
- Faculty of Medicine, University of Lisbon, 1649-028 Lisbon, Portugal
| | - Carolina Pestana
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
- Centre of Statistics and Its Applications, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
| | - Ana C. Queirós
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | - Cristina João
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
- Faculty of Medical Sciences, NOVA Medical School, 1169-056 Lisbon, Portugal
- Hemato-Oncology Department of Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | - Emilie Arnault Carneiro
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
| |
Collapse
|
19
|
Sun J, Park C, Guenthner N, Gurley S, Zhang L, Lubben B, Adebayo O, Bash H, Chen Y, Maksimos M, Muz B, Azab AK. Tumor-associated macrophages in multiple myeloma: advances in biology and therapy. J Immunother Cancer 2022; 10:e003975. [PMID: 35428704 PMCID: PMC9014078 DOI: 10.1136/jitc-2021-003975] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2022] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma (MM) is a cancer of plasma cells in the bone marrow (BM) and represents the second most common hematological malignancy in the world. The MM tumor microenvironment (TME) within the BM niche consists of a wide range of elements which play important roles in supporting MM disease progression, survival, proliferation, angiogenesis, as well as drug resistance. Together, the TME fosters an immunosuppressive environment in which immune recognition and response are repressed. Macrophages are a central player in the immune system with diverse functions, and it has been long established that macrophages play a critical role in both inducing direct and indirect immune responses in cancer. Tumor-associated macrophages (TAMs) are a major population of cells in the tumor site. Rather than contributing to the immune response against tumor cells, TAMs in many cancers are found to exhibit protumor properties including supporting chemoresistance, tumor proliferation and survival, angiogenesis, immunosuppression, and metastasis. Targeting TAM represents a novel strategy for cancer immunotherapy, which has potential to indirectly stimulate cytotoxic T cell activation and recruitment, and synergize with checkpoint inhibitors and chemotherapies. In this review, we will provide an updated and comprehensive overview into the current knowledge on the roles of TAMs in MM, as well as the therapeutic targets that are being explored as macrophage-targeted immunotherapy, which may hold key to future therapeutics against MM.
Collapse
Affiliation(s)
- Jennifer Sun
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University in St. Louis McKelvey School of Engineering, St. Louis, Missouri, USA
| | - Chaelee Park
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Nicole Guenthner
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Shannon Gurley
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Luna Zhang
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University in St. Louis McKelvey School of Engineering, St. Louis, Missouri, USA
| | - Berit Lubben
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Ola Adebayo
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Hannah Bash
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Yixuan Chen
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Mina Maksimos
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Barbara Muz
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Abdel Kareem Azab
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University in St. Louis McKelvey School of Engineering, St. Louis, Missouri, USA
| |
Collapse
|
20
|
Goodarzi A, Valikhani M, Amiri F, Safari A. The mechanisms of mutual relationship between malignant hematologic cells and mesenchymal stem cells: Does it contradict the nursing role of mesenchymal stem cells? Cell Commun Signal 2022; 20:21. [PMID: 35236376 PMCID: PMC8889655 DOI: 10.1186/s12964-022-00822-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/18/2021] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are known as the issue in biology because of some unpredictable characteristics in the different microenvironments especially in their bone marrow niche. MSCs are used in the regenerative medicine because of their unique potentials for trans-differentiation, immunomodulation, and paracrine capacity. But, their pathogenic and pro-survival effects in tumors/cancers including hematologic malignancies are indisputable. MSCs and/or their derivatives might be involved in tumor growth, metastasis and drug resistance in the leukemias. One of important relationship is MSCs and hematologic malignancy-derived cells which affects markedly the outcome of disease. The communication between these two cells may be contact-dependent and/or contact-independent. In this review, we studied the crosstalk between MSCs and malignant hematologic cells which results the final feedback either the progression or suppression of blood cell malignancy. Video abstract.
Collapse
Affiliation(s)
- Alireza Goodarzi
- Department of Medical Laboratory Sciences, School of Paramedicine, Hamadan University of Medical Sciences, Shahid Fahmideh Blvd., The Opposite Side of Mardom Park, Hamadan, 6517838741, Iran
| | - Mohsen Valikhani
- Hematology Department, School of Allied Medical Science, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Amiri
- Department of Medical Laboratory Sciences, School of Paramedicine, Hamadan University of Medical Sciences, Shahid Fahmideh Blvd., The Opposite Side of Mardom Park, Hamadan, 6517838741, Iran.
| | - Armita Safari
- Student Research Committee, Hamadan University of Medical Science, Hamadan, Iran
| |
Collapse
|
21
|
Bone marrow sinusoidal endothelium controls terminal erythroid differentiation and reticulocyte maturation. Nat Commun 2021; 12:6963. [PMID: 34845225 PMCID: PMC8630019 DOI: 10.1038/s41467-021-27161-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 10/28/2021] [Indexed: 12/19/2022] Open
Abstract
Within the bone marrow microenvironment, endothelial cells (EC) exert important functions. Arterial EC support hematopoiesis while H-type capillaries induce bone formation. Here, we show that BM sinusoidal EC (BM-SEC) actively control erythropoiesis. Mice with stabilized β-catenin in BM-SEC (Ctnnb1OE-SEC) generated by using a BM-SEC-restricted Cre mouse line (Stab2-iCreF3) develop fatal anemia. While activation of Wnt-signaling in BM-SEC causes an increase in erythroblast subsets (PII-PIV), mature erythroid cells (PV) are reduced indicating impairment of terminal erythroid differentiation/reticulocyte maturation. Transplantation of Ctnnb1OE-SEC hematopoietic stem cells into wildtype recipients confirms lethal anemia to be caused by cell-extrinsic, endothelial-mediated effects. Ctnnb1OE-SEC BM-SEC reveal aberrant sinusoidal differentiation with altered EC gene expression and perisinusoidal ECM deposition and angiocrine dysregulation with de novo endothelial expression of FGF23 and DKK2, elevated in anemia and involved in vascular stabilization, respectively. Our study demonstrates that BM-SEC play an important role in the bone marrow microenvironment in health and disease.
Collapse
|
22
|
Resistance to Tyrosine Kinase Inhibitors in Chronic Myeloid Leukemia-From Molecular Mechanisms to Clinical Relevance. Cancers (Basel) 2021; 13:cancers13194820. [PMID: 34638304 PMCID: PMC8508378 DOI: 10.3390/cancers13194820] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 01/18/2023] Open
Abstract
Simple Summary Chronic myeloid leukemia (CML) is a myeloproliferative neoplasia associated with a molecular alteration, the fusion gene BCR-ABL1, that encodes the tyrosine kinase oncoprotein BCR-ABL1. This led to the development of tyrosine kinase inhibitors (TKI), with Imatinib being the first TKI approved. Although the vast majority of CML patients respond to Imatinib, resistance to this targeted therapy contributes to therapeutic failure and relapse. Here we review the molecular mechanisms and other factors (e.g., patient adherence) involved in TKI resistance, the methodologies to access these mechanisms, and the possible therapeutic approaches to circumvent TKI resistance in CML. Abstract Resistance to targeted therapies is a complex and multifactorial process that culminates in the selection of a cancer clone with the ability to evade treatment. Chronic myeloid leukemia (CML) was the first malignancy recognized to be associated with a genetic alteration, the t(9;22)(q34;q11). This translocation originates the BCR-ABL1 fusion gene, encoding the cytoplasmic chimeric BCR-ABL1 protein that displays an abnormally high tyrosine kinase activity. Although the vast majority of patients with CML respond to Imatinib, a tyrosine kinase inhibitor (TKI), resistance might occur either de novo or during treatment. In CML, the TKI resistance mechanisms are usually subdivided into BCR-ABL1-dependent and independent mechanisms. Furthermore, patients’ compliance/adherence to therapy is critical to CML management. Techniques with enhanced sensitivity like NGS and dPCR, the use of artificial intelligence (AI) techniques, and the development of mathematical modeling and computational prediction methods could reveal the underlying mechanisms of drug resistance and facilitate the design of more effective treatment strategies for improving drug efficacy in CML patients. Here we review the molecular mechanisms and other factors involved in resistance to TKIs in CML and the new methodologies to access these mechanisms, and the therapeutic approaches to circumvent TKI resistance.
Collapse
|
23
|
Mannino G, Russo C, Maugeri G, Musumeci G, Vicario N, Tibullo D, Giuffrida R, Parenti R, Lo Furno D. Adult stem cell niches for tissue homeostasis. J Cell Physiol 2021; 237:239-257. [PMID: 34435361 PMCID: PMC9291197 DOI: 10.1002/jcp.30562] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/26/2021] [Accepted: 08/09/2021] [Indexed: 12/13/2022]
Abstract
Adult stem cells are fundamental to maintain tissue homeostasis, growth, and regeneration. They reside in specialized environments called niches. Following activating signals, they proliferate and differentiate into functional cells that are able to preserve tissue physiology, either to guarantee normal turnover or to counteract tissue damage caused by injury or disease. Multiple interactions occur within the niche between stem cell‐intrinsic factors, supporting cells, the extracellular matrix, and signaling pathways. Altogether, these interactions govern cell fate, preserving the stem cell pool, and regulating stem cell proliferation and differentiation. Based on their response to body needs, tissues can be largely classified into three main categories: tissues that even in normal conditions are characterized by an impressive turnover to replace rapidly exhausting cells (blood, epidermis, or intestinal epithelium); tissues that normally require only a basal cell replacement, though able to efficiently respond to increased tissue needs, injury, or disease (skeletal muscle); tissues that are equipped with less powerful stem cell niches, whose repairing ability is not able to overcome severe damage (heart or nervous tissue). The purpose of this review is to describe the main characteristics of stem cell niches in these different tissues, highlighting the various components influencing stem cell activity. Although much has been done, more work is needed to further increase our knowledge of niche interactions. This would be important not only to shed light on this fundamental chapter of human physiology but also to help the development of cell‐based strategies for clinical therapeutic applications, especially when other approaches fail.
Collapse
Affiliation(s)
- Giuliana Mannino
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Cristina Russo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Grazia Maugeri
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Nunzio Vicario
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Daniele Tibullo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Rosario Giuffrida
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Debora Lo Furno
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
24
|
Gu W, Qu R, Meng F, Cornelissen JJLM, Zhong Z. Polymeric nanomedicines targeting hematological malignancies. J Control Release 2021; 337:571-588. [PMID: 34364920 DOI: 10.1016/j.jconrel.2021.08.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/31/2021] [Accepted: 08/02/2021] [Indexed: 10/20/2022]
Abstract
Hematological malignancies (HMs) typically persisting in the blood, lymphoma, and/or bone marrow invalidate surgery and local treatments clinically used for solid tumors. The presence and drug resistance nature of cancer stem cells (CSCs) further lends HMs hard to cure. The development of new treatments like molecular targeted drugs and antibodies has improved the clinical outcomes for HMs but only to a certain extent, due to issues of low bioavailability, moderate response, occurrence of drug resistance, and/or dose-limiting toxicities. In the past years, polymeric nanomedicines targeting HMs including refractory and relapsed lymphoma, leukemia and multiple myeloma have emerged as a promising chemotherapeutic approach that is shown capable of overcoming drug resistance, delivering drugs not only to cancer cells but also CSCs, and increasing therapeutic index by lessening drug-associated adverse effects. In addition, polymeric nanomedicines have shown to potentiate next-generation anticancer modalities such as therapeutic proteins and nucleic acids in effectively treating HMs. In this review, we highlight recent advance in targeted polymeric nanoformulations that are coated with varying ligands (e.g. cancer cell membrane proteins, antibodies, transferrin, hyaluronic acid, aptamer, peptide, and folate) and loaded with different therapeutic agents (e.g. chemotherapeutics, molecular targeted drugs, therapeutic antibodies, nucleic acid drugs, and apoptotic proteins) for directing to distinct targets (e.g. CD19, CD20, CD22, CD30, CD38, CD44, CD64, CXCR, FLT3, VLA-4, and bone marrow microenvironment) in HMs. The advantages and potential challenges of different designs are discussed.
Collapse
Affiliation(s)
- Wenxing Gu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; Department of Biomolecular Nanotechnology, MESA+ Institute for Nanotechnology, University of Twente, 7500 AE Enschede, the Netherlands
| | - Ruobing Qu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China.
| | - Jeroen J L M Cornelissen
- Department of Biomolecular Nanotechnology, MESA+ Institute for Nanotechnology, University of Twente, 7500 AE Enschede, the Netherlands.
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|
25
|
Lemus-Conejo A, Medrano M, Lopez S, Millan-Linares MC, Rosillo MA, Perez-Simon JA, Muriana FJG, Abia R. MUFAs in High-Fat Diets Protect against Obesity-Induced Bias of Hematopoietic Cell Lineages. Mol Nutr Food Res 2021; 65:e2001203. [PMID: 34132459 DOI: 10.1002/mnfr.202001203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 05/27/2021] [Indexed: 11/08/2022]
Abstract
SCOPE The role of dietary fatty acids in the generation of bone marrow (BM) immune cells and their trafficking to extramedullary compartments in the obesity is not yet fully understood. METHODS AND RESULTS C57BL/6J mice are randomly assigned to isocaloric high-fat diets (HFDs) formulate with dietary fats rich in saturated fatty acids (SFAs), monounsaturated fatty acids (MUFAs) or MUFAs fortified with eicosapentaenoic and docosahexaenoic acids for 20 weeks, followed by profiling of the obese metabolic phenotype and immunophenotypic features of immune cells in blood, spleen, and BM. All HFDs induce an obese phenotype, but it becomes largely less disruptive after the HFDs are enriched in MUFAs, which also induce signs of granulopoiesis and an expansion of long-term hematopoietic stem and granulocyte-macrophage progenitor cells in BM. In contrast, a HFD enriched in SFAs disturbs the fitness of medullary lymphocytes and promotes monopoiesis in favor of pro-inflammatory activated subsets. CONCLUSION The reshaping of the fatty acid pools with MUFAs from the diet serves to manipulate the generation and trafficking of immune cells that are biased during obesity. These findings reveal a novel strategy by which dietary MUFAs may be instrumental in combating HFD-induced dysfunctional immune systems.
Collapse
Affiliation(s)
- Ana Lemus-Conejo
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, The Spanish National Research Council (CSIC), Seville, 41013, Spain
| | - Mayte Medrano
- Department of Haematology, Instituto de Biomedicina de Sevilla (IBiS/CSIC/CIBERONC), Hospital Universitario Virgen del Rocio, University of Seville, Seville, 41012, Spain
| | - Sergio Lopez
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, The Spanish National Research Council (CSIC), Seville, 41013, Spain
- Department of Cell Biology, Faculty of Biology, University of Seville, Seville, 41012, Spain
- Instituto de Biomedicina de Sevilla (IBiS/CSIC), Hospital Universitario Virgen del Rocio, University of Seville, Seville, 41012, Spain
| | | | - Maria A Rosillo
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, The Spanish National Research Council (CSIC), Seville, 41013, Spain
| | - Jose A Perez-Simon
- Department of Haematology, Instituto de Biomedicina de Sevilla (IBiS/CSIC/CIBERONC), Hospital Universitario Virgen del Rocio, University of Seville, Seville, 41012, Spain
| | - Francisco J G Muriana
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, The Spanish National Research Council (CSIC), Seville, 41013, Spain
| | - Rocio Abia
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, The Spanish National Research Council (CSIC), Seville, 41013, Spain
| |
Collapse
|
26
|
Sun G, Shvab A, Leclerc GJ, Li B, Beckedorff F, Shiekhattar R, Barredo JC. Protein Kinase D-Dependent Downregulation of Immediate Early Genes through Class IIA Histone Deacetylases in Acute Lymphoblastic Leukemia. Mol Cancer Res 2021; 19:1296-1307. [PMID: 33980612 DOI: 10.1158/1541-7786.mcr-20-0808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 03/12/2021] [Accepted: 05/04/2021] [Indexed: 11/16/2022]
Abstract
Acute lymphoblastic leukemia (ALL) is a leading cause of cancer-related death in children and adolescents, and cure rates for relapsed/refractory ALL remain dismal, highlighting the need for novel targeted therapies. To identify genome-wide metabolic-stress regulated genes, we used RNA-sequencing in ALL cells treated with AICAR, an AMPK activator. RNA-sequencing identified the immediate early genes (IEGs) as a subset of genes downregulated by AICAR. We show that AICAR-induced IEGs downregulation was blocked by an adenosine uptake inhibitor indicating AICAR was responsible for IEGs reprogramming. Using pharmacologic and genetic models we established this mechanism was AMPK-independent. Further investigations using kinase assays, PKD/PKC inhibitors and rescue experiments, demonstrated that AICAR directly inhibited PKD kinase activity and identified PKD as responsible for IEGs downregulation. Mechanistically, PKD inhibition suppressed phosphorylation and nuclear export of class IIa HDACs, which lowered histone H3 acetylation and decreased NFκB(p65) recruitment to IEGs promoters. Finally, PKD inhibition induced apoptosis via DUSP1/DUSP6 downregulation eliciting a DNA damage response. More importantly, ALL patient cells exhibited the same PKD-HDACs-IEGs-mediated mechanism. As proof of principle of the therapeutic potential of targeting PKD, we established the in vivo relevance of our findings using an NSG ALL mouse model. In conclusion, we identified a previously unreported PKD-dependent survival mechanism in response to AICAR-induced cellular stress in ALL through regulation of DUSPs and IEGs' expression. IMPLICATIONS: PKD mediates early transcriptional responses in ALL cells as an adaptive survival mechanism to overcome cellular stress.
Collapse
Affiliation(s)
- Guangyan Sun
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
| | - Anna Shvab
- Cancer Biology Program, University of Miami Miller School of Medicine, Miami, Florida
| | - Guy J Leclerc
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
| | - Bin Li
- Stemsynergy Therapeutics, Inc, Miami, Florida
| | - Felipe Beckedorff
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida
| | - Ramin Shiekhattar
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida.,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Julio C Barredo
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida. .,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida.,Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida.,Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
27
|
Abstract
Hematopoiesis is the process that leads to multiple leukocyte lineage generation within the bone marrow. This process is maintained throughout life thanks to a nonstochastic division of hematopoietic stem cells (HSCs), where during each division, one daughter cell retains pluripotency while the other differentiates into a restricted multipotent progenitor (MPP) that converts into mature, committed circulating cell. This process is tightly regulated at the level of cellular metabolism and the shift from anaerobic glycolysis, typical of quiescent HSC, to oxidative metabolism fosters HSCs proliferation and commitment. Systemic and local factors influencing metabolism alter HSCs balance under pathological conditions, with chronic metabolic and inflammatory diseases driving HSCs commitment toward activated blood immune cell subsets. This is the case of atherosclerosis, where impaired systemic lipid metabolism affects HSCs epigenetics that reflects into increased differentiation toward activated circulating subsets. Aim of this review is to discuss the impact of lipids and lipoproteins on HSCs pathophysiology, with a focus on the molecular mechanisms influencing cellular metabolism. A better understanding of these aspects will shed light on innovative strategies to target atherosclerosis-associated inflammation.
Collapse
|
28
|
Pastorczak A, Domka K, Fidyt K, Poprzeczko M, Firczuk M. Mechanisms of Immune Evasion in Acute Lymphoblastic Leukemia. Cancers (Basel) 2021; 13:1536. [PMID: 33810515 PMCID: PMC8037152 DOI: 10.3390/cancers13071536] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/17/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) results from a clonal expansion of abnormal lymphoid progenitors of B cell (BCP-ALL) or T cell (T-ALL) origin that invade bone marrow, peripheral blood, and extramedullary sites. Leukemic cells, apart from their oncogene-driven ability to proliferate and avoid differentiation, also change the phenotype and function of innate and adaptive immune cells, leading to escape from the immune surveillance. In this review, we provide an overview of the genetic heterogeneity and treatment of BCP- and T-ALL. We outline the interactions of leukemic cells in the bone marrow microenvironment, mainly with mesenchymal stem cells and immune cells. We describe the mechanisms by which ALL cells escape from immune recognition and elimination by the immune system. We focus on the alterations in ALL cells, such as overexpression of ligands for various inhibitory receptors, including anti-phagocytic receptors on macrophages, NK cell inhibitory receptors, as well as T cell immune checkpoints. In addition, we describe how developing leukemia shapes the bone marrow microenvironment and alters the function of immune cells. Finally, we emphasize that an immunosuppressive microenvironment can reduce the efficacy of chemo- and immunotherapy and provide examples of preclinical studies showing strategies for improving ALL treatment by targeting these immunosuppressive interactions.
Collapse
Affiliation(s)
- Agata Pastorczak
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, 91-738 Lodz, Poland;
| | - Krzysztof Domka
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (K.D.); (K.F.); (M.P.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Klaudyna Fidyt
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (K.D.); (K.F.); (M.P.)
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Martyna Poprzeczko
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (K.D.); (K.F.); (M.P.)
| | - Malgorzata Firczuk
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (K.D.); (K.F.); (M.P.)
| |
Collapse
|
29
|
Lana JF, da Fonseca LF, Azzini G, Santos G, Braga M, Cardoso Junior AM, Murrell WD, Gobbi A, Purita J, Percope de Andrade MA. Bone Marrow Aspirate Matrix: A Convenient Ally in Regenerative Medicine. Int J Mol Sci 2021; 22:ijms22052762. [PMID: 33803231 PMCID: PMC7963152 DOI: 10.3390/ijms22052762] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/17/2021] [Accepted: 02/24/2021] [Indexed: 02/06/2023] Open
Abstract
The rise in musculoskeletal disorders has prompted medical experts to devise novel effective alternatives to treat complicated orthopedic conditions. The ever-expanding field of regenerative medicine has allowed researchers to appreciate the therapeutic value of bone marrow-derived biological products, such as the bone marrow aspirate (BMA) clot, a potent orthobiologic which has often been dismissed and regarded as a technical complication. Numerous in vitro and in vivo studies have contributed to the expansion of medical knowledge, revealing optimistic results concerning the application of autologous bone marrow towards various impactful disorders. The bone marrow accommodates a diverse family of cell populations and a rich secretome; therefore, autologous BMA-derived products such as the “BMA Matrix”, may represent a safe and viable approach, able to reduce the costs and some drawbacks linked to the expansion of bone marrow. BMA provides —it eliminates many hurdles associated with its preparation, especially in regards to regulatory compliance. The BMA Matrix represents a suitable alternative, indicated for the enhancement of tissue repair mechanisms by modulating inflammation and acting as a natural biological scaffold as well as a reservoir of cytokines and growth factors that support cell activity. Although promising, more clinical studies are warranted in order to further clarify the efficacy of this strategy.
Collapse
Affiliation(s)
- José Fábio Lana
- IOC—Instituto do Osso e da Cartilagem, 1386 Presidente Kennedy Avenue, Indaiatuba 13334-170, Brazil; (J.F.L.); (G.A.)
| | | | - Gabriel Azzini
- IOC—Instituto do Osso e da Cartilagem, 1386 Presidente Kennedy Avenue, Indaiatuba 13334-170, Brazil; (J.F.L.); (G.A.)
| | - Gabriel Santos
- IOC—Instituto do Osso e da Cartilagem, 1386 Presidente Kennedy Avenue, Indaiatuba 13334-170, Brazil; (J.F.L.); (G.A.)
- Correspondence:
| | - Marcelo Braga
- Hospital São Judas Tadeu, 150 Cel. João Notini St, Divinópolis 35500-017, Brazil;
| | - Alvaro Motta Cardoso Junior
- Núcleo Avançado de Estudos em Ortopedia e Neurocirurgia, 2144 Ibirapuera Avenue, São Paulo 04028-001, Brazil;
| | - William D. Murrell
- Abu Dhabi Knee and Sports Medicine, Healthpoint Hospital, Zayed Sports City, Between Gate 1 and 6, Abu Dhabi 00000 (P. O. Box No. 112308), United Arab Emirates;
- 411th Hospital Center, Bldg 938, Birmingham Ave, Naval Air Station, Jacksonville, FL 32212, USA
| | - Alberto Gobbi
- O.A.S.I. Bioresearch Foundation Gobbi Onlus, 20133 Milano, Italy;
| | - Joseph Purita
- Institute of Regenerative Medicine, Boca Raton, FL 33432, USA;
| | | |
Collapse
|
30
|
Minciacchi VR, Kumar R, Krause DS. Chronic Myeloid Leukemia: A Model Disease of the Past, Present and Future. Cells 2021; 10:cells10010117. [PMID: 33435150 PMCID: PMC7827482 DOI: 10.3390/cells10010117] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 12/31/2020] [Accepted: 01/07/2021] [Indexed: 12/11/2022] Open
Abstract
Chronic myeloid leukemia (CML) has been a "model disease" with a long history. Beginning with the first discovery of leukemia and the description of the Philadelphia Chromosome and ending with the current goal of achieving treatment-free remission after targeted therapies, we describe here the journey of CML, focusing on molecular pathways relating to signaling, metabolism and the bone marrow microenvironment. We highlight current strategies for combination therapies aimed at eradicating the CML stem cell; hopefully the final destination of this long voyage.
Collapse
MESH Headings
- Epigenesis, Genetic
- History, 20th Century
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/history
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Models, Biological
- Molecular Targeted Therapy
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Tumor Microenvironment/genetics
Collapse
Affiliation(s)
- Valentina R. Minciacchi
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Str. 42-44, 60596 Frankfurt am Main, Germany; (V.R.M.); (R.K.)
| | - Rahul Kumar
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Str. 42-44, 60596 Frankfurt am Main, Germany; (V.R.M.); (R.K.)
| | - Daniela S. Krause
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Str. 42-44, 60596 Frankfurt am Main, Germany; (V.R.M.); (R.K.)
- German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
- Frankfurt Cancer Institute, 60596 Frankfurt, Germany
- Faculty of Medicine, Medical Clinic II, Johann Wolfgang Goethe University, 60596 Frankfurt, Germany
- Correspondence: ; Tel.: +49-69-63395-500; Fax: +49-69-63395-519
| |
Collapse
|
31
|
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
|
32
|
Hou D, Wang B, You R, Wang X, Liu J, Zhan W, Chen P, Qin T, Zhang X, Huang H. Stromal cells promote chemoresistance of acute myeloid leukemia cells via activation of the IL-6/STAT3/OXPHOS axis. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1346. [PMID: 33313091 PMCID: PMC7723653 DOI: 10.21037/atm-20-3191] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background Bone marrow stromal cells (BMSCs) are known to promote chemoresistance in acute myeloid leukemia (AML) cells. However, the molecular basis for BMSC-associated AML chemoresistance remains largely unexplored. Methods The mitochondrial oxidative phosphorylation (OXPHOS) levels of AML cells were measured by a Seahorse XFe24 cell metabolic analyzer. The activity of total or mitochondrial signal transducer and transcription activator 3 (STAT3) in AML cells was explored by flow cytometry and Western blotting. Real-time quantitative PCR, Western blotting and enzyme-linked immunosorbent assay (ELISA) were used to analyze expression of interleukin 6 (IL-6) in the human BMSC line HS-5, and IL-6 was knocked out in HS-5 cells by CRISPR/Cas9 system. Results In this study, we observed that co-culturing with BMSCs heightened OXPHOS levels in AML cells, thus promoting chemoresistance in these cells. HS-5 cell-induced upregulation of OXPHOS is dependent on the activation of STAT3, especially on that of mitochondrial serine phosphorylated STAT3 (pS-STAT3) in AML cells. The relationship among pS-STAT3, OXPHOS, and chemosensitivity of AML cells induced by BMSCs was demonstrated by the STAT3 activator and inhibitor, which upregulated and downregulated the levels of mitochondrial pS-STAT3 and OXPHOS, respectively. Intriguingly, AML cells remodeled HS-5 cells to secrete more IL-6, which augmented mitochondrial OXPHOS in AML cells and stimulated their chemoresistance. IL-6 knockout in HS-5 cells impaired the ability of these cells to activate STAT3, to increase OXPHOS, or to promote chemoresistance in AML cells. Conclusions BMSCs promoted chemoresistance in AML cells via the activation of the IL-6/STAT3/OXPHOS pathway. These findings exhibit a novel mechanism of chemoresistance in AML cells in the bone marrow microenvironment from a metabolic perspective.
Collapse
Affiliation(s)
- Diyu Hou
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Bin Wang
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China.,Clinical Laboratory, Fujian Children's Hospital, Fuzhou, China
| | - Ruolan You
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaoting Wang
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jingru Liu
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Weiwu Zhan
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ping Chen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Tiandi Qin
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xuehao Zhang
- School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, China
| | - Huifang Huang
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
33
|
Bone marrow aspirate clot: A feasible orthobiologic. J Clin Orthop Trauma 2020; 11:S789-S794. [PMID: 32999557 PMCID: PMC7503156 DOI: 10.1016/j.jcot.2020.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 07/05/2020] [Indexed: 12/29/2022] Open
Abstract
Musculoskeletal disorders are one of the major health burdens and a leading source of disability worldwide, affecting both juvenile and elderly populations either as a consequence of ageing or extrinsic factors such as physical injuries. This condition often involves a group of locomotor structures such as the bones, joints and muscles and may therefore cause significant economic and emotional impact. Some pharmacological and non-pharmacological treatments have been considered as potential solutions, however, these alternatives have provided quite limited efficacy due to the short-term effect on pain management and inability to restore damaged tissue. The emergence of novel therapeutic alternatives such as the application of orthobiologics, particularly bone marrow aspirate (BMA) clot, have bestowed medical experts with considerable optimism as evidenced by the significant results found in numerous studies addressed in this manuscript. Although other products have been proposed for the treatment of musculoskeletal injuries, the peculiar interest in BMA, fibrin clot and associated fibrinolytic mechanisms continues to expand. BMA is a rich source of various cellular and molecular components which have demonstrated positive effects on tissue regeneration in many in vitro and in vivo models of musculoskeletal injuries. In addition to being able to undergo self-renewal and differentiation, the hematopoietic and mesenchymal stem cells present in this orthobiologic elicit key immunomodulatory and paracrine roles in inflammatory responses in tissue injury and drive the coagulation cascade towards tissue repair via different mechanisms. Although promising, these complex regenerative mechanisms have not yet been fully elucidated.
Collapse
|
34
|
Kurian CJ, Thomas C, Houtmann S, Klumpp T, Binder AF. Case Report: Concomitant Diagnosis of Plasma Cell Leukemia in Patient With JAK2 Positive Myeloproliferative Neoplasm. Front Oncol 2020; 10:1497. [PMID: 32974181 PMCID: PMC7482648 DOI: 10.3389/fonc.2020.01497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/13/2020] [Indexed: 11/27/2022] Open
Abstract
Plasma cell dyscrasias and myeloproliferative neoplasms (MPN) are hematologic malignancies arising from two distinct hematopoietic cell lineages. They rarely occur concomitantly. Here, we report a case of a patient with a recent diagnosis of a JAK2 V617F positive MPN who presented with a new diagnosis of plasma cell leukemia. The patient had presented to the hospital with a leukocytosis predominantly comprised of plasma cells, followed by work-up involving peripheral blood flow cytometry, FISH analysis, and bone-marrow biopsy. FISH analysis was suggestive of a common progenitor cell for these distinct hematologic malignancies. To our knowledge, this case represents the second reported instance of a concomitant JAK2 positive MPN with primary plasma cell leukemia.
Collapse
Affiliation(s)
- Christine J Kurian
- Department of Internal Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, United States
| | - Colin Thomas
- Department of Internal Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, United States
| | - Sarah Houtmann
- Department of Internal Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, United States
| | - Thomas Klumpp
- Department of Medical Oncology, Thomas Jefferson University Hospital, Philadelphia, PA, United States
| | - Adam Finn Binder
- Department of Medical Oncology, Thomas Jefferson University Hospital, Philadelphia, PA, United States
| |
Collapse
|
35
|
A Review of the Action of Magnesium on Several Processes Involved in the Modulation of Hematopoiesis. Int J Mol Sci 2020; 21:ijms21197084. [PMID: 32992944 PMCID: PMC7582682 DOI: 10.3390/ijms21197084] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/17/2020] [Accepted: 09/22/2020] [Indexed: 12/18/2022] Open
Abstract
Magnesium (Mg2+) is an essential mineral for the functioning and maintenance of the body. Disturbances in Mg2+ intracellular homeostasis result in cell-membrane modification, an increase in oxidative stress, alteration in the proliferation mechanism, differentiation, and apoptosis. Mg2+ deficiency often results in inflammation, with activation of inflammatory pathways and increased production of proinflammatory cytokines by immune cells. Immune cells and others that make up the blood system are from hematopoietic tissue in the bone marrow. The hematopoietic tissue is a tissue with high indices of renovation, and Mg2+ has a pivotal role in the cell replication process, as well as DNA and RNA synthesis. However, the impact of the intra- and extracellular disturbance of Mg2+ homeostasis on the hematopoietic tissue is little explored. This review deals specifically with the physiological requirements of Mg2+ on hematopoiesis, showing various studies related to the physiological requirements and the effects of deficiency or excess of this mineral on the hematopoiesis regulation, as well as on the specific process of erythropoiesis, granulopoiesis, lymphopoiesis, and thrombopoiesis. The literature selected includes studies in vitro, in animal models, and in humans, giving details about the impact that alterations of Mg2+ homeostasis can have on hematopoietic cells and hematopoietic tissue.
Collapse
|
36
|
Kindlin-3 loss curbs chronic myeloid leukemia in mice by mobilizing leukemic stem cells from protective bone marrow niches. Proc Natl Acad Sci U S A 2020; 117:24326-24335. [PMID: 32929022 DOI: 10.1073/pnas.2009078117] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Kindlin-3 (K3)-mediated integrin adhesion controls homing and bone marrow (BM) retention of normal hematopoietic cells. However, the role of K3 in leukemic stem cell (LSC) retention and growth in the remodeled tumor-promoting BM is unclear. We report that loss of K3 in a mouse model of chronic myeloid leukemia (CML) triggers the release of LSCs from the BM into the circulation and impairs their retention, proliferation, and survival in secondary organs, which curbs CML development, progression, and metastatic dissemination. We found de novo expression of cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) on CML-LSCs but not normal hematopoietic stem cells and this enabled us to specifically deplete K3 with a CTLA-4-binding RNA aptamer linked to a K3-siRNA (small interfering RNA) in CTLA-4+ LSCs in vivo, which mobilized LSCs in the BM, induced disease remission, and prolonged survival of mice with CML. Thus, disrupting interactions of LSCs with the BM environment is a promising strategy to halt the disease-inducing and relapse potential of LSCs.
Collapse
|
37
|
Clara-Trujillo S, Gallego Ferrer G, Gómez Ribelles JL. In Vitro Modeling of Non-Solid Tumors: How Far Can Tissue Engineering Go? Int J Mol Sci 2020; 21:E5747. [PMID: 32796596 PMCID: PMC7460836 DOI: 10.3390/ijms21165747] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/07/2020] [Accepted: 08/10/2020] [Indexed: 12/19/2022] Open
Abstract
In hematological malignancies, leukemias or myelomas, malignant cells present bone marrow (BM) homing, in which the niche contributes to tumor development and drug resistance. BM architecture, cellular and molecular composition and interactions define differential microenvironments that govern cell fate under physiological and pathological conditions and serve as a reference for the native biological landscape to be replicated in engineered platforms attempting to reproduce blood cancer behavior. This review summarizes the different models used to efficiently reproduce certain aspects of BM in vitro; however, they still lack the complexity of this tissue, which is relevant for fundamental aspects such as drug resistance development in multiple myeloma. Extracellular matrix composition, material topography, vascularization, cellular composition or stemness vs. differentiation balance are discussed as variables that could be rationally defined in tissue engineering approaches for achieving more relevant in vitro models. Fully humanized platforms closely resembling natural interactions still remain challenging and the question of to what extent accurate tissue complexity reproduction is essential to reliably predict drug responses is controversial. However, the contributions of these approaches to the fundamental knowledge of non-solid tumor biology, its regulation by niches, and the advance of personalized medicine are unquestionable.
Collapse
Affiliation(s)
- Sandra Clara-Trujillo
- Center for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, 46022 Valencia, Spain; (G.G.F.); (J.L.G.R.)
- Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 46022 Valencia, Spain
| | - Gloria Gallego Ferrer
- Center for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, 46022 Valencia, Spain; (G.G.F.); (J.L.G.R.)
- Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 46022 Valencia, Spain
| | - José Luis Gómez Ribelles
- Center for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, 46022 Valencia, Spain; (G.G.F.); (J.L.G.R.)
- Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 46022 Valencia, Spain
| |
Collapse
|
38
|
Extracellular Adenine Nucleotides and Adenosine Modulate the Growth and Survival of THP-1 Leukemia Cells. Int J Mol Sci 2020; 21:ijms21124425. [PMID: 32580317 PMCID: PMC7352165 DOI: 10.3390/ijms21124425] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/19/2020] [Accepted: 06/20/2020] [Indexed: 12/26/2022] Open
Abstract
A new approach to improve the effectiveness of acute myeloid leukemia (AML) treatment is to use the properties of purinergic signaling molecules secreted into the bone marrow milieu in response to leukemic cell growth. Therefore, our study aimed to evaluate the effects of extracellular adenine nucleotides and adenosine on the growth and death parameters in the leukemic THP-1 cell line. Cells were exposed to ATP, ADP, AMP, adenosine and nonhydrolyzable analogues of ATP and ADP (ATPγS and ADPβS) in a 1–1000 μM broad concentration range. The basal mRNA expression of the P1 and P2 receptors was evaluated by real-time PCR. Changes in the processes of cell growth and death were assessed by flow cytometry analysis of proliferation, cell cycle and apoptosis. Chemotaxis toward stromal cell-derived factor-1 (SDF-1) was performed using the modified Boyden chamber assay, and chemokine receptor type 4 (CXCR4) surface expression was quantified by flow cytometry. We indicated several antileukemic actions. High micromolar concentrations (100–1000 μM) of extracellular adenine nucleotides and adenosine inhibit the growth of cells by arresting the cell cycle and/or inducing apoptosis. ATP is characterized by the highest potency and widest range of effects, and is responsible for the cell cycle arrest and the apoptosis induction. Compared to ATP, the effect of ADP is slightly weaker. Adenosine mostly has a cytotoxic effect, with the induction of apoptosis. The last studied nucleotide, AMP, demonstrated only a weak cytotoxic effect without affecting the cell cycle. In addition, cell migration towards SDF-1 was inhibited by low micromolar concentrations (10 μM). One of the reasons for this action of ATPγS and adenosine was a reduction in CXCR4 surface expression, but this only partially explains the mechanism of antimigratory action. In summary, extracellular adenine nucleotides and adenosine inhibit THP-1 cell growth, cause death of cells and modulate the functioning of the SDF-1/CXCR4 axis. Thus, they negatively affect the processes that are responsible for the progression of AML and the difficulties in AML treatment.
Collapse
|
39
|
Role of the Bone Marrow Milieu in Multiple Myeloma Progression and Therapeutic Resistance. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:e752-e768. [PMID: 32651110 DOI: 10.1016/j.clml.2020.05.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/22/2020] [Accepted: 05/29/2020] [Indexed: 01/10/2023]
Abstract
Multiple myeloma (MM) is a cancer of the plasma cells within the bone marrow (BM). Studies have shown that the cellular and noncellular components of the BM milieu, such as cytokines and exosomes, play an integral role in MM pathogenesis and progression by mediating drug resistance and inducing MM proliferation. Moreover, the BM microenvironment of patients with MM facilitates cancer tolerance and immune evasion through the expansion of regulatory immune cells, inhibition of antitumor effector cells, and disruption of the antigen presentation machinery. These are of special relevance, especially in the current era of cancer immunotherapy. An improved understanding of the supportive role of the MM BM microenvironment will allow for the development of future therapies targeting MM in the context of the BM milieu to elicit deeper and more durable responses. In the present review, we have discussed our current understanding of the role of the BM microenvironment in MM progression and resistance to therapy and discuss novel potential approaches to alter its pro-MM function.
Collapse
|
40
|
Krüger T, Middeke JM, Stölzel F, Mütherig A, List C, Brandt K, Heidrich K, Teipel R, Ordemann R, Schuler U, Oelschlägel U, Wermke M, Kräter M, Herbig M, Wehner R, Schmitz M, Bornhäuser M, von Bonin M. Reliable isolation of human mesenchymal stromal cells from bone marrow biopsy specimens in patients after allogeneic hematopoietic cell transplantation. Cytotherapy 2019; 22:21-26. [PMID: 31883948 DOI: 10.1016/j.jcyt.2019.10.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/25/2019] [Accepted: 10/28/2019] [Indexed: 12/28/2022]
Abstract
Isolation of mesenchymal stromal cells (MSCs) from pretreated, hematologic patients is challenging. Especially after allogeneic hematopoietic cell transplantation (HCT), standard protocols using bone marrow aspirates fail to reliably recover sufficient cell numbers. Because MSCs are considered to contribute to processes that mainly affect the outcome after transplantation, such as an efficient lymphohematopoietic recovery, extent of graft-versus-host disease as well as the occurrence of leukemic relapse, it is of great clinical relevance to investigate MSC function in this context. Previous studies showed that MSCs can be isolated by collagenase digestion of large bone fragments of hematologically healthy patients undergoing hip replacement or knee surgeries. We have now further developed this procedure for the isolation of MSCs from hematologic patients after allogeneic HCT by using trephine biopsy specimens obtained during routine examinations. Comparison of aspirates and trephine biopsy specimens from patients after allogeneic HCT revealed a significantly higher frequency of clonogenic MSCs (colony-forming unit-fibroblast [CFU-F]) in trephine biopsy specimens (mean, 289.8 ± standard deviation 322.5 CFU-F colonies/1 × 106 total nucleated cells versus 4.2 ± 9.9; P < 0.0001). Subsequent expansion of functional MSCs isolated from trephine biopsy specimen was more robust and led to a significantly higher yield compared with control samples expanded from aspirates (median, 1.6 × 106; range, 0-2.3 × 107 P0 MSCs versus 5.4 × 104; range, 0-8.9 × 106; P < 0.0001). Using trephine biopsy specimens as MSC source facilitates the investigation of various clinical questions.
Collapse
Affiliation(s)
- Thomas Krüger
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Jan Moritz Middeke
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Friedrich Stölzel
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Anke Mütherig
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Catrin List
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Kalina Brandt
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Katharina Heidrich
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Raphael Teipel
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Rainer Ordemann
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Ulrich Schuler
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Uta Oelschlägel
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Martin Wermke
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany; University Cancer Centrum (UCC), Early Clinical Trial Unit (ECTU), University Hospital Carl Gustav Carus, Dresden, Germany
| | - Martin Kräter
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Maik Herbig
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany; Biotechnology Center, Center for Molecular and Cellular Bioengineering TU Dresden Tatzberg 47-49, Dresden, Germany
| | - Rebekka Wehner
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
| | - Marc Schmitz
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany; Center for Regenerative Therapies (CRTD), Dresden, Germany
| | - Martin Bornhäuser
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany; Center for Regenerative Therapies (CRTD), Dresden, Germany
| | - Malte von Bonin
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
41
|
Sai B, Dai Y, Fan S, Wang F, Wang L, Li Z, Tang J, Wang L, Zhang X, Zheng L, Chen F, Li G, Xiang J. Cancer-educated mesenchymal stem cells promote the survival of cancer cells at primary and distant metastatic sites via the expansion of bone marrow-derived-PMN-MDSCs. Cell Death Dis 2019; 10:941. [PMID: 31819035 PMCID: PMC6901580 DOI: 10.1038/s41419-019-2149-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 11/12/2019] [Indexed: 12/21/2022]
Abstract
Bone marrow mesenchymal stem cells (BMSCs) are multipotent stromal cells that can differentiate into a variety of cell types. BMSCs are chemotactically guided towards the cancer cells and contribute to the formation of a cancer microenvironment. The homing of BMSCs was affected by various factors. Disseminated tumour cells (DTCs) in distant organs, especially in the bone marrow, are the source of cancer metastasis and cancer relapse. DTC survival is also determined by the microenvironment. Here we aim to elucidate how cancer-educated BMSCs promote the survival of cancer cells at primary tumour sites and distant sites. We highlight the dynamic change by identifying different gene expression signatures in intratumoral BMSCs and in BMSCs that move back in the bone marrow. Intratumoral BMSCs acquire high mobility and displayed immunosuppressive effects. Intratumoral BMSCs that ultimately home to the bone marrow exhibit a strong immunosuppressive function. Cancer-educated BMSCs promote the survival of lung cancer cells via expansion of MDSCs in bone marrow, primary tumour sites and metastatic sites. These Ly6G+ MDSCs suppress proliferation of T cells. CXCL5, nitric oxide and GM-CSF produced by cancer-educated BMSCs contribute to the formation of malignant microenvironments. Treatment with CXCL5 antibody, the iNOS inhibitor 1400w and GM-CSF antibody reduced MDSC expansion in the bone marrow, primary tumour sites and metastatic sites, and promoted the efficiency of PD-L1 antibody. Our study reveals that cancer-educated BMSCs are the component of the niche for primary lung cancer cells and DTCs, and that they can be the target for immunotherapy.
Collapse
Affiliation(s)
- Buqing Sai
- NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, Hunan, 410013, China
| | - Yafei Dai
- NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, Hunan, 410013, China
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Fan Wang
- NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, Hunan, 410013, China
| | - Lujuan Wang
- NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, Hunan, 410013, China
| | - Zheng Li
- NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Jingqun Tang
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Li Wang
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Xina Zhang
- NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, Hunan, 410013, China
| | - Leliang Zheng
- NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, Hunan, 410013, China
| | - Fei Chen
- Department of Spinal Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, Hunan, 410013, China
| | - Juanjuan Xiang
- NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China.
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, Hunan, 410013, China.
| |
Collapse
|
42
|
Spertini C, Baïsse B, Bellone M, Gikic M, Smirnova T, Spertini O. Acute Myeloid and Lymphoblastic Leukemia Cell Interactions with Endothelial Selectins: Critical Role of PSGL-1, CD44 and CD43. Cancers (Basel) 2019; 11:cancers11091253. [PMID: 31461905 PMCID: PMC6770432 DOI: 10.3390/cancers11091253] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 08/22/2019] [Accepted: 08/23/2019] [Indexed: 12/30/2022] Open
Abstract
Acute myeloid and lymphoblastic leukemia are poor prognosis hematologic malignancies, which disseminate from the bone marrow into the blood. Blast interactions with selectins expressed by vascular endothelium promote the development of drug resistance and leukostasis. While the role of selectins in initiating leukemia blast adhesion is established, our knowledge of the involved selectin ligands is incomplete. Using various primary acute leukemia cells and U937 monoblasts, we identified here functional selectin ligands expressed by myeloblasts and lymphoblasts by performing biochemical studies, expression inhibition by RNA interference and flow adhesion assays on recombinant selectins or selectin ligands immunoadsorbed from primary blast cells. Results demonstrate that P-selectin glycoprotein ligand-1 (PSGL-1) is the major P-selectin ligand on myeloblasts, while it is much less frequently expressed and used by lymphoblasts to interact with endothelial selectins. To roll on E-selectin, myeloblasts use PSGL-1, CD44, and CD43 to various extents and the contribution of these ligands varies strongly among patients. In contrast, the interactions of PSGL-1-deficient lymphoblasts with E-selectin are mainly supported by CD43 and/or CD44. By identifying key selectin ligands expressed by acute leukemia blasts, this study offers novel insight into their involvement in mediating acute leukemia cell adhesion with vascular endothelium and may identify novel therapeutic targets.
Collapse
Affiliation(s)
- Caroline Spertini
- Service and Central Laboratory of Hematology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Bénédicte Baïsse
- Service and Central Laboratory of Hematology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Marta Bellone
- Service and Central Laboratory of Hematology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Milica Gikic
- Service and Central Laboratory of Hematology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Tatiana Smirnova
- Service and Central Laboratory of Hematology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Olivier Spertini
- Service and Central Laboratory of Hematology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, CH-1011 Lausanne, Switzerland.
| |
Collapse
|
43
|
Constructing Three-Dimensional Microenvironments Using Engineered Biomaterials for Hematopoietic Stem Cell Expansion. TISSUE ENGINEERING PART B-REVIEWS 2019; 25:312-329. [DOI: 10.1089/ten.teb.2018.0286] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
44
|
Gnani D, Crippa S, della Volpe L, Rossella V, Conti A, Lettera E, Rivis S, Ometti M, Fraschini G, Bernardo ME, Di Micco R. An early-senescence state in aged mesenchymal stromal cells contributes to hematopoietic stem and progenitor cell clonogenic impairment through the activation of a pro-inflammatory program. Aging Cell 2019; 18:e12933. [PMID: 30828977 PMCID: PMC6516180 DOI: 10.1111/acel.12933] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 01/22/2019] [Accepted: 02/02/2019] [Indexed: 12/14/2022] Open
Abstract
Hematopoietic stem and progenitor cells (HSPC) reside in the bone marrow (BM) niche and serve as a reservoir for mature blood cells throughout life. Aging in the BM is characterized by low‐grade chronic inflammation that could contribute to the reduced functionality of aged HSPC. Mesenchymal stromal cells (MSC) in the BM support HSPC self‐renewal. However, changes in MSC function with age and the crosstalk between MSC and HSPC remain understudied. Here, we conducted an extensive characterization of senescence features in BM‐derived MSC from young and aged healthy donors. Aged MSC displayed an enlarged senescent‐like morphology, a delayed clonogenic potential and reduced proliferation ability when compared to younger counterparts. Of note, the observed proliferation delay was associated with increased levels of SA‐β‐galactosidase (SA‐β‐Gal) and lipofuscin in aged MSC at early passages and a modest but consistent accumulation of physical DNA damage and DNA damage response (DDR) activation. Consistent with the establishment of a senescence‐like state in aged MSC, we detected an increase in pro‐inflammatory senescence‐associated secretory phenotype (SASP) factors, both at the transcript and protein levels. Conversely, the immunomodulatory properties of aged MSC were significantly reduced. Importantly, exposure of young HSPC to factors secreted by aged MSC induced pro‐inflammatory genes in HSPC and impaired HSPC clonogenic potential in a SASP‐dependent manner. Altogether, our results reveal that BM‐derived MSC from aged healthy donors display features of senescence and that, during aging, MSC‐associated secretomes contribute to activate an inflammatory transcriptional program in HSPC that may ultimately impair their functionality.
Collapse
Affiliation(s)
- Daniela Gnani
- San Raffaele Telethon Institute for Gene Therapy Milan Italy
| | - Stefania Crippa
- San Raffaele Telethon Institute for Gene Therapy Milan Italy
| | - Lucrezia della Volpe
- San Raffaele Telethon Institute for Gene Therapy Milan Italy
- Vita‐Salute San Raffaele University Milan Italy
| | | | - Anastasia Conti
- San Raffaele Telethon Institute for Gene Therapy Milan Italy
| | - Emanuele Lettera
- San Raffaele Telethon Institute for Gene Therapy Milan Italy
- Vita‐Salute San Raffaele University Milan Italy
| | - Silvia Rivis
- San Raffaele Telethon Institute for Gene Therapy Milan Italy
| | - Marco Ometti
- Department of Orthopedics and Traumatology San Raffaele Hospital Scientific Institute Milan Italy
| | - Gianfranco Fraschini
- Department of Orthopedics and Traumatology San Raffaele Hospital Scientific Institute Milan Italy
| | - Maria Ester Bernardo
- San Raffaele Telethon Institute for Gene Therapy Milan Italy
- Pediatric Immunohematology and Bone Marrow Transplantation Unit San Raffaele Scientific Institute Milan Italy
| | | |
Collapse
|
45
|
Comparison of Hematopoietic and Spermatogonial Stem Cell Niches from the Regenerative Medicine Aspect. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1107:15-40. [PMID: 29882209 DOI: 10.1007/5584_2018_217] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|