1
|
Guo X, Fu Y, Peng J, Fu Y, Dong S, Ding RB, Qi X, Bao J. Emerging anticancer potential and mechanisms of snake venom toxins: A review. Int J Biol Macromol 2024; 269:131990. [PMID: 38704067 DOI: 10.1016/j.ijbiomac.2024.131990] [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: 11/27/2023] [Revised: 03/13/2024] [Accepted: 04/28/2024] [Indexed: 05/06/2024]
Abstract
Animal-derived venom, like snake venom, has been proven to be valuable natural resources for the drug development. Previously, snake venom was mainly investigated in its pharmacological activities in regulating coagulation, vasodilation, and cardiovascular function, and several marketed cardiovascular drugs were successfully developed from snake venom. In recent years, snake venom fractions have been demonstrated with anticancer properties of inducing apoptotic and autophagic cell death, restraining proliferation, suppressing angiogenesis, inhibiting cell adhesion and migration, improving immunity, and so on. A number of active anticancer enzymes and peptides have been identified from snake venom toxins, such as L-amino acid oxidases (LAAOs), phospholipase A2 (PLA2), metalloproteinases (MPs), three-finger toxins (3FTxs), serine proteinases (SPs), disintegrins, C-type lectin-like proteins (CTLPs), cell-penetrating peptides, cysteine-rich secretory proteins (CRISPs). In this review, we focus on summarizing these snake venom-derived anticancer components on their anticancer activities and underlying mechanisms. We will also discuss their potential to be developed as anticancer drugs in the future.
Collapse
Affiliation(s)
- Xijun Guo
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Yuanfeng Fu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Junbo Peng
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Ying Fu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Shuai Dong
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Ren-Bo Ding
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Xingzhu Qi
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China.
| | - Jiaolin Bao
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China.
| |
Collapse
|
2
|
Fatima N, Shen Y, Crassini K, Burling O, Thurgood L, Iwanowicz EJ, Lang H, Karanewsky DS, Christopherson RI, Mulligan SP, Best OG. The CIpP activator, TR-57, is highly effective as a single agent and in combination with venetoclax against CLL cells in vitro. Leuk Lymphoma 2024; 65:585-597. [PMID: 38227293 DOI: 10.1080/10428194.2023.2300055] [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: 10/24/2022] [Accepted: 12/11/2023] [Indexed: 01/17/2024]
Abstract
Despite advances in treatment, a significant proportion of patients with chronic lymphocytic leukemia (CLL) will relapse with drug-resistant disease. The imipridones, ONC-201 and ONC-212, are effective against a range of different cancers, including acute myeloid leukemia (AML) and tumors of the brain, breast, and prostate. These drugs induce cell death through activation of the mitochondrial protease, caseinolytic protease (CIpP), and the unfolded protein response (UPR). Here we demonstrate that the novel imipridone analog, TR-57, has efficacy as a single agent and synergises with venetoclax against CLL cells under in vitro conditions that mimic the tumor microenvironment. Changes in protein expression suggest TR-57 activates the UPR, inhibits the AKT and ERK1/2 pathways and induces pro-apoptotic changes in the expression of proteins of the BCL-2 family. The study suggests that TR-57, as a single agent and in combination with venetoclax, may represent an effective treatment option for CLL.
Collapse
MESH Headings
- Humans
- Sulfonamides/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Apoptosis/drug effects
- Drug Synergism
- Cell Line, Tumor
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Unfolded Protein Response/drug effects
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Proto-Oncogene Proteins c-bcl-2/genetics
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Narjis Fatima
- Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, Camperdown, Australia
- School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
| | - Yandong Shen
- Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, Camperdown, Australia
- School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
| | - Kyle Crassini
- Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, Camperdown, Australia
| | - Olivia Burling
- Flinders Health and Medical Research Institute, Department of Genetics and Molecular Medicine, College of Medicine and Public Health, Flinders University, Camperdown, Australia
| | - Lauren Thurgood
- Flinders Health and Medical Research Institute, Department of Genetics and Molecular Medicine, College of Medicine and Public Health, Flinders University, Camperdown, Australia
| | | | - Henk Lang
- Madera Therapeutics, LLC, Cary, North Carolina, USA
| | | | | | - Stephen P Mulligan
- Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, Camperdown, Australia
- School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
| | - O Giles Best
- Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, Camperdown, Australia
- School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
- Flinders Health and Medical Research Institute, Department of Genetics and Molecular Medicine, College of Medicine and Public Health, Flinders University, Camperdown, Australia
| |
Collapse
|
3
|
Zhang X, Lei Y, Chen X, He J, Liu Z, Zhu W, Xu Y, Jin X. Suppression of NSCLC progression via the co-administration of Danusertib, an AURK inhibitor, and KRIBB11, an HSF1 inhibitor. Biochem Pharmacol 2024; 223:116155. [PMID: 38521474 DOI: 10.1016/j.bcp.2024.116155] [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: 08/15/2023] [Revised: 12/27/2023] [Accepted: 03/19/2024] [Indexed: 03/25/2024]
Abstract
Aurora kinase (AURK) and heat shock factor 1 (HSF1) are commonly overexpressed in non-small cell lung cancer (NSCLC), correlating with poor prognosis. This study aims to assess the therapeutic potential of combining the Danusertib (Danu, AURK inhibitor) and KRIBB11 (HSF1 inhibitor) for NSCLC treatment. The effects of this combination were investigated in A549 cells and a tumor xenograft mouse model. The findings demonstrate that concurrent administration of Danu and KRIBB11 effectively impedes cell proliferation, induces apoptosis, and triggers G2/M cell cycle arrest. Moreover, the combination treatment upregulates pro-apoptotic proteins (Cleaved-caspase3, Cleaved-PARP, and Bax) while downregulating anti-apoptotic proteins (Bcl-2), as well as G2/M-related proteins (CDC2 and cyclin B1). Additionally, the combination treatment elevates reactive oxygen species (ROS) levels, decreases mitochondrial membrane potential, and activates the DNA damage pathway. Interestingly, we discovered that the PI3K/AKT pathway is involved in mediating the effects of both Danu and KRIBB11. Furthermore, the combination treatment inhibits tumor growth and AKT signaling in the xenograft mouse model, increases levels of the tumor tissue oxidation product malondialdehyde (MDA), and induces DNA damage. To summarize, a potential therapeutic approach for NSCLC may involve dual inhibition of AURK and HSF1, resulting in the downregulation of the PI3K/AKT signaling pathway, and the activation of ROS-mediated mitochondrial and DNA damage pathways.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China; Department of Thoracic Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang Province 324000, China
| | - Ying Lei
- Department of Respiratory and Critical Care Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Ouzhou, Zhejiang Province 324000, China
| | - Xiang Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China
| | - Jiahuang He
- Department of Respiratory and Critical Care Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Ouzhou, Zhejiang Province 324000, China
| | - Zitian Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China
| | - Wentao Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China
| | - Yi Xu
- Department of Science & Technology, Department of Urology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Ouzhou, Zhejiang Province 324000, China.
| | - Xuru Jin
- Department of Respiratory and Critical Care Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Ouzhou, Zhejiang Province 324000, China; Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China.
| |
Collapse
|
4
|
Mouawad N, Ruggeri E, Capasso G, Martinello L, Visentin A, Frezzato F, Trentin L. How receptor tyrosine kinase-like orphan receptor 1 meets its partners in chronic lymphocytic leukemia. Hematol Oncol 2024; 42:e3250. [PMID: 38949887 DOI: 10.1002/hon.3250] [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: 10/12/2023] [Revised: 12/29/2023] [Accepted: 01/02/2024] [Indexed: 07/03/2024]
Abstract
Chronic lymphocytic leukemia (CLL) is the most common leukemia in western societies, recognized by clinical and molecular heterogeneity. Despite the success of targeted therapies, acquired resistance remains a challenge for relapsed and refractory CLL, as a consequence of mutations in the target or the upregulation of other survival pathways leading to the progression of the disease. Research on proteins that can trigger such pathways may define novel therapies for a successful outcome in CLL such as the receptor tyrosine kinase-like orphan receptor 1 (ROR1). ROR1 is a signaling receptor for Wnt5a, with an important role during embryogenesis. The aberrant expression on CLL cells and several types of tumors, is involved in cell proliferation, survival, migration as well as drug resistance. Antibody-based immunotherapies and small-molecule compounds emerged to target ROR1 in preclinical and clinical studies. Efforts have been made to identify new prognostic markers having predictive value to refine and increase the detection and management of CLL. ROR1 can be considered as an attractive target for CLL diagnosis, prognosis, and treatment. It can be clinically effective alone and/or in combination with current approved agents. In this review, we summarize the scientific achievements in targeting ROR1 for CLL diagnosis, prognosis, and treatment.
Collapse
MESH Headings
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Humans
- Receptor Tyrosine Kinase-like Orphan Receptors/metabolism
- Prognosis
- Molecular Targeted Therapy
- Animals
- Biomarkers, Tumor/metabolism
Collapse
Affiliation(s)
- Nayla Mouawad
- Hematology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Edoardo Ruggeri
- Hematology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Guido Capasso
- Hematology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Leonardo Martinello
- Hematology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Andrea Visentin
- Hematology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Federica Frezzato
- Hematology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Livio Trentin
- Hematology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| |
Collapse
|
5
|
Choi JW, Kim SW, Kim HS, Kang MJ, Kim SA, Han JY, Kim H, Ku SY. Effects of Melatonin, GM-CSF, IGF-1, and LIF in Culture Media on Embryonic Development: Potential Benefits of Individualization. Int J Mol Sci 2024; 25:751. [PMID: 38255823 PMCID: PMC10815572 DOI: 10.3390/ijms25020751] [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: 11/21/2023] [Revised: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
The implantation of good-quality embryos to the receptive endometrium is essential for successful live birth through in vitro fertilization (IVF). The higher the quality of embryos, the higher the live birth rate per cycle, and so efforts have been made to obtain as many high-quality embryos as possible after fertilization. In addition to an effective controlled ovarian stimulation process to obtain high-quality embryos, the composition of the embryo culture medium in direct contact with embryos in vitro is also important. During embryonic development, under the control of female sex hormones, the fallopian tubes and endometrium create a microenvironment that supplies the nutrients and substances necessary for embryos at each stage. During this process, the development of the embryo is finely regulated by signaling molecules, such as growth factors and cytokines secreted from the epithelial cells of the fallopian tube and uterine endometrium. The development of embryo culture media has continued since the first successful human birth through IVF in 1978. However, there are still limitations to mimicking a microenvironment similar to the reproductive organs of women suitable for embryo development in vitro. Efforts have been made to overcome the harsh in vitro culture environment and obtain high-quality embryos by adding various supplements, such as antioxidants and growth factors, to the embryo culture medium. Recently, there has been an increase in the number of studies on the effect of supplementation in different clinical situations such as old age, recurrent implantation failure (RIF), and unexplained infertility; in addition, anticipation of the potential benefits from individuation is rising. This article reviews the effects of representative supplements in culture media on embryo development.
Collapse
Affiliation(s)
- Jung-Won Choi
- Laboratory of In Vitro Fertilization, Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea; (J.-W.C.); (H.-S.K.); (M.-J.K.); (S.-A.K.)
| | - Sung-Woo Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea; (S.-W.K.); (J.-Y.H.); (H.K.)
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hee-Sun Kim
- Laboratory of In Vitro Fertilization, Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea; (J.-W.C.); (H.-S.K.); (M.-J.K.); (S.-A.K.)
| | - Moon-Joo Kang
- Laboratory of In Vitro Fertilization, Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea; (J.-W.C.); (H.-S.K.); (M.-J.K.); (S.-A.K.)
| | - Sung-Ah Kim
- Laboratory of In Vitro Fertilization, Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea; (J.-W.C.); (H.-S.K.); (M.-J.K.); (S.-A.K.)
| | - Ji-Yeon Han
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea; (S.-W.K.); (J.-Y.H.); (H.K.)
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hoon Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea; (S.-W.K.); (J.-Y.H.); (H.K.)
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Seung-Yup Ku
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea; (S.-W.K.); (J.-Y.H.); (H.K.)
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul 03080, Republic of Korea
| |
Collapse
|
6
|
Dunn KA, MacDonald E, MacDonald T, Kulkarni K. Bacterial heat shock protein genes during induction chemotherapy in pediatric patients with acute lymphoblastic leukemia. Future Oncol 2024; 20:17-23. [PMID: 38189148 DOI: 10.2217/fon-2023-0263] [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] [Indexed: 01/09/2024] Open
Abstract
Background: Heat shock proteins (HSP) protect cancer cells. Gastrointestinal bacteria contain HSP genes and can release extracellular vesicles which act as biological shuttles. Stress from treatment may result in a microbial community with more HSP genes, which could contribute to circulating HSP levels. Methods: The authors examined the abundance of five bacterial HSP genes pre-treatment and during induction in stool sequences from 30 pediatric acute lymphoblastic leukemia patients. Results: Decreased mean HTPG counts (p = 0.0024) pre-treatment versus induction were observed. During induction, HTPG, Shannon diversity and Bacteroidetes decreased (p = 7.5e-4; 1.1e-3; 8.6e-4), while DNAK and Firmicutes increased (p = 6.9e-3; 9.2e-4). Conclusion: Understanding microbial HSP gene community changes with treatment is the first step in determining if bacterial HSPs are important to the tumor microenvironment and leukemia treatment.
Collapse
Affiliation(s)
- Katherine A Dunn
- Department of Pediatrics, Division of Hematology Oncology, Izaak Walton Killam (IWK) Health, Halifax, NS, Canada
- Department of Biology, Dalhousie University, Halifax, NS, Canada
- Institute for Comparative Genomics, Dalhousie University, Halifax, NS, Canada
| | - Emma MacDonald
- Department of Pediatrics, Division of Hematology Oncology, Izaak Walton Killam (IWK) Health, Halifax, NS, Canada
| | - Tamara MacDonald
- Department of Pharmacy, IWK Health, Halifax, NS, Canada
- Faculty of Health Professions, Dalhousie University, Halifax, NS, Canada
| | - Ketan Kulkarni
- Department of Pediatrics, Division of Hematology Oncology, Izaak Walton Killam (IWK) Health, Halifax, NS, Canada
| |
Collapse
|
7
|
Sklavenitis-Pistofidis R, Lightbody ED, Reidy M, Tsuji J, Aranha MP, Heilpern-Mallory D, Huynh D, Chong SJF, Hackett L, Haradhvala NJ, Wu T, Su NK, Berrios B, Alberge JB, Dutta A, Davids MS, Papaioannou M, Getz G, Ghobrial IM, Manier S. Systematic characterization of therapeutic vulnerabilities in Multiple Myeloma with Amp1q reveals increased sensitivity to the combination of MCL1 and PI3K inhibitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.01.551480. [PMID: 37577538 PMCID: PMC10418223 DOI: 10.1101/2023.08.01.551480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
The development of targeted therapy for patients with Multiple Myeloma (MM) is hampered by the low frequency of actionable genetic abnormalities. Gain or amplification of chr1q (Amp1q) is the most frequent arm-level copy number gain in patients with MM, and it is associated with higher risk of progression and death despite recent advances in therapeutics. Thus, developing targeted therapy for patients with MM and Amp1q stands to benefit a large portion of patients in need of more effective management. Here, we employed large-scale dependency screens and drug screens to systematically characterize the therapeutic vulnerabilities of MM with Amp1q and showed increased sensitivity to the combination of MCL1 and PI3K inhibitors. Using single-cell RNA sequencing, we compared subclones with and without Amp1q within the same patient tumors and showed that Amp1q is associated with higher levels of MCL1 and the PI3K pathway. Furthermore, by isolating isogenic clones with different copy number for part of the chr1q arm, we showed increased sensitivity to MCL1 and PI3K inhibitors with arm-level gain. Lastly, we demonstrated synergy between MCL1 and PI3K inhibitors and dissected their mechanism of action in MM with Amp1q.
Collapse
Affiliation(s)
- Romanos Sklavenitis-Pistofidis
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Elizabeth D. Lightbody
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Mairead Reidy
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Junko Tsuji
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Michelle P. Aranha
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Daniel Heilpern-Mallory
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Daisy Huynh
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Stephen J. F. Chong
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Liam Hackett
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nicholas J. Haradhvala
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Ting Wu
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Nang K. Su
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Brianna Berrios
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jean-Baptiste Alberge
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Ankit Dutta
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Matthew S. Davids
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Maria Papaioannou
- Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Hematology Unit, 1st Internal Medicine Department, AHEPA University Hospital, Thessaloniki, Greece
| | - Gad Getz
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Irene M. Ghobrial
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Salomon Manier
- INSERM UMRS1277, CNRS UMR9020, Lille University, 59000, France
- Department of Hematology, CHU Lille, Lille University, 59000, France
| |
Collapse
|
8
|
Wong SHD, Yin B, Li Z, Yuan W, Zhang Q, Xie X, Tan Y, Wong N, Zhang K, Bian L. Mechanical manipulation of cancer cell tumorigenicity via heat shock protein signaling. SCIENCE ADVANCES 2023; 9:eadg9593. [PMID: 37418519 PMCID: PMC10328411 DOI: 10.1126/sciadv.adg9593] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 06/01/2023] [Indexed: 07/09/2023]
Abstract
Biophysical cues of rigid tumor matrix play a critical role in cancer cell malignancy. We report that stiffly confined cancer cells exhibit robust growth of spheroids in the stiff hydrogel that exerts substantial confining stress on the cells. The stressed condition activated Hsp (heat shock protein)-signal transducer and activator of transcription 3 signaling via the transient receptor potential vanilloid 4-phosphatidylinositol 3-kinase/Akt axis, thereby up-regulating the expression of the stemness-related markers in cancer cells, whereas these signaling activities were suppressed in cancer cells cultured in softer hydrogels or stiff hydrogels with stress relief or Hsp70 knockdown/inhibition. This mechanopriming based on three-dimensional culture enhanced cancer cell tumorigenicity and metastasis in animal models upon transplantation, and pharmaceutically inhibiting Hsp70 improved the anticancer efficacy of chemotherapy. Mechanistically, our study reveals the crucial role of Hsp70 in regulating cancer cell malignancy under mechanically stressed conditions and its impacts on cancer prognosis-related molecular pathways for cancer treatments.
Collapse
Affiliation(s)
- Siu Hong Dexter Wong
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, P. R. China
- Research Institute for Sports Science and Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, P. R. China
| | - Bohan Yin
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, P. R. China
| | - Zhuo Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong 999077, P. R. China
| | - Weihao Yuan
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong 999077, P. R. China
| | - Qin Zhang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, P. R. China
| | - Xian Xie
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong 999077, P. R. China
| | - Youhua Tan
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, P. R. China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, P. R. China
| | - Nathalie Wong
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong 999077, P. R. China
| | - Kunyu Zhang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P. R. China
| | - Liming Bian
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P. R. China
| |
Collapse
|
9
|
Mouawad N, Capasso G, Ruggeri E, Martinello L, Severin F, Visentin A, Facco M, Trentin L, Frezzato F. Is It Still Possible to Think about HSP70 as a Therapeutic Target in Onco-Hematological Diseases? Biomolecules 2023; 13:biom13040604. [PMID: 37189352 DOI: 10.3390/biom13040604] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/21/2023] [Accepted: 03/26/2023] [Indexed: 03/30/2023] Open
Abstract
The search for molecules to be targeted that are involved in apoptosis resistance/increased survival and pathogenesis of onco-hematological malignancies is ongoing since these diseases are still not completely understood. Over the years, a good candidate has been identified in the Heat Shock Protein of 70kDa (HSP70), a molecule defined as “the most cytoprotective protein ever been described”. HSP70 is induced in response to a wide variety of physiological and environmental insults, allowing cells to survive lethal conditions. This molecular chaperone has been detected and studied in almost all the onco-hematological diseases and is also correlated to poor prognosis and resistance to therapy. In this review, we give an overview of the discoveries that have led us to consider HSP70 as a therapeutic target for mono- or combination-therapies in acute and chronic leukemias, multiple myeloma and different types of lymphomas. In this excursus, we will also consider HSP70 partners, such as its transcription factor HSF1 or its co-chaperones whose druggability could indirectly affect HSP70. Finally, we will try to answer the question asked in the title of this review considering that, despite the effort made by research in this field, HSP70 inhibitors never reached the clinic.
Collapse
|
10
|
Heat-Shock Proteins in Leukemia and Lymphoma: Multitargets for Innovative Therapeutic Approaches. Cancers (Basel) 2023; 15:cancers15030984. [PMID: 36765939 PMCID: PMC9913431 DOI: 10.3390/cancers15030984] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/08/2023] Open
Abstract
Heat-shock proteins (HSPs) are powerful chaperones that provide support for cellular functions under stress conditions but also for the homeostasis of basic cellular machinery. All cancer cells strongly rely on HSPs, as they must continuously adapt to internal but also microenvironmental stresses to survive. In solid tumors, HSPs have been described as helping to correct the folding of misfolded proteins, sustain oncogenic pathways, and prevent apoptosis. Leukemias and lymphomas also overexpress HSPs, which are frequently associated with resistance to therapy. HSPs have therefore been proposed as new therapeutic targets. Given the specific biology of hematological malignancies, it is essential to revise their role in this field, providing a more adaptable and comprehensive picture that would help design future clinical trials. To that end, this review will describe the different pathways and functions regulated by HSP27, HSP70, HSP90, and, not least, HSP110 in leukemias and lymphomas.
Collapse
|
11
|
He YF, Hu XM, Khan MA, Yu BY, Sheng YC, Xiao XZ, Wan XX, Tan SP, Xiong K. HSF1 Alleviates Brain Injury by Inhibiting NLRP3-Induced Pyroptosis in a Sepsis Model. Mediators Inflamm 2023; 2023:2252255. [PMID: 36741074 PMCID: PMC9897924 DOI: 10.1155/2023/2252255] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/15/2022] [Accepted: 09/16/2022] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Sepsis, which could cause a systemic inflammatory response, is a life-threatening disease with a high morbidity and mortality rate. There is evidence that brain injury may be related to severe systemic infection induced by sepsis. The brain injury caused by sepsis could increase the risk of mortality in septic patients, which seriously affects the septic patient's prognosis of survival. Although there remains a focus on sepsis research, clinical measures to prevent and treat brain injury in sepsis are not yet available, and the high mortality rate is still a big health burden. Therefore, it is necessary to investigate the new molecules or regulated pathways that can effectively inhibit the progress of sepsis. OBJECTIVE NLR family pyrin domain-containing 3 (NLRP3) increased in the procession of sepsis and functioned as the key regulator of pyroptosis. Heat shock factor 1 (HSF1) can protect organs from multiorgan dysfunction syndrome induced by lipopolysaccharides in mice, and NLRP3 could be inhibited by HSF1 in many organs. However, whether HSF1 regulated NLRP3 in sepsis-induced brain injury, as well as the detailed mechanism of HSF1 in brain injury, remains unknown in the sepsis model. In this research, we try to explore the relationship between HSF1 and NLRP3 in a sepsis model and try to reveal the mechanism of HSF1 inhibiting the process of brain injury. METHODS In this study, we used wild-type mice and hsf1 -/- mice for in vivo research and PC12 cells for in vitro research. Real-time PCR and Western blot were used to analyze the expression of HSF1, NLRP3, cytokines, and pyrolytic proteins. EthD-III staining was chosen to detect the pyroptosis of the hippocampus and PC12 cells. RESULTS The results showed that HSF1 is negatively related to pyroptosis. The pyroptosis in cells of brain tissue was significantly increased in the hsf1 -/- mouse model compared to hsf1 +/+ mice. In PC12 cells, hsf1 siRNA can upregulate pyroptosis while HSF1-transfected plasmid could inhibit the pyroptosis. HSF1 could negatively regulate the NLRP3 pathway in PC12 cells, while hsf1 siRNA enhanced the pyroptosis in PC12 cells, which could be reversed by nlrp3 siRNA. CONCLUSION These results imply that HSF1 could alleviate sepsis-induced brain injury by inhibiting pyroptosis through the NLRP3-dependent pathway in brain tissue and PC12 cells, suggesting HSF1 as a potential molecular target for treating brain injury in sepsis clinical studies.
Collapse
Affiliation(s)
- Yi-fu He
- Department of Obstetrics and Gynecology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xi-min Hu
- Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Md. Asaduzzaman Khan
- The Research Centre for Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Bo-yao Yu
- Clinical Medicine Five-Year Program, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Yi-cun Sheng
- Clinical Medicine Five-Year Program, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Xian-zhong Xiao
- Key Laboratory of Sepsis Translational Medicine of Hunan, Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Xin-xing Wan
- Department of Endocrinology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Si-pin Tan
- Key Laboratory of Sepsis Translational Medicine of Hunan, Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, China
- Hunan Key Laboratory of Ophthalmology, Changsha 410008, China
- Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| |
Collapse
|
12
|
Wang Y, Zhu Q, Guo S, Ao J, Zhang W, Fei J, Yu S, Niu M, Zhang Y, Sherman MY, Xiao ZXJ, Yi Y. HSF1 activates the FOXO3a-ΔNp63α-CDK4 axis to promote head and neck squamous cell carcinoma cell proliferation and tumour growth. FEBS Lett 2023; 597:1125-1137. [PMID: 36700826 DOI: 10.1002/1873-3468.14588] [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: 11/01/2022] [Revised: 01/01/2023] [Accepted: 01/11/2023] [Indexed: 01/27/2023]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is one of the most prevalent cancers worldwide. Heat shock factor 1 (HSF1) is a conserved transcriptional factor that plays a critical role in maintaining cellular proteostasis. However, the role of HSF1 in HNSCC development remains largely unclear. Here, we report that HSF1 promotes forkhead box protein O3a (FOXO3a)-dependent transcription of ΔNp63α (p63 isoform in the p53 family; inhibits cell migration, invasion, and metastasis), which leads to upregulation of cyclin-dependent kinase 4 expression and HNSCC tumour growth. Ablation of HSF1 or treatment with KRIBB11, a specific pharmacological inhibitor of HSF1, significantly suppresses ΔNp63α expression and HNSCC tumour growth. Clinically, the expression of HSF1 is positively correlated with the expression of ΔNp63α in HNSCC tumours. Together, this study demonstrates that the HSF1-ΔNp63α pathway is critically important for HNSCC tumour growth.
Collapse
Affiliation(s)
- Yuemeng Wang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Qile Zhu
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Shiya Guo
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Juan Ao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Wenhua Zhang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Junjie Fei
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Shuhan Yu
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Mengmeng Niu
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yujun Zhang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | | | - Zhi-Xiong Jim Xiao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Yong Yi
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Lyu C, Wang Q, Yin X, Li Z, Wang T, Wang Y, Cui S, Liu K, Wang Z, Gao C, Xu R. Clinical significance and potential mechanism of heat shock factor 1 in acute myeloid leukemia. Aging (Albany NY) 2022; 14:7026-7037. [PMID: 36069792 PMCID: PMC9512492 DOI: 10.18632/aging.204267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 08/17/2022] [Indexed: 11/25/2022]
Abstract
Background: Heat shock factor 1 (HSF1) is now considered to have the potential to be used as a prognostic biomarker in cancers. However, its clinical significance and potential function in acute myeloid leukemia (AML) remain underexplored. Methods: In this study, the expression pattern and clinical significance of HSF1 in AML were examined by integrating data from databases including The Cancer Genome Atlas (TCGA), The Genotype–Tissue Expression (GTEx), Vizome, Cancer Cell Line Encyclopedia (CCLE) and Gene Expression Omnibus (GEO). Linkedomics was applied to collect HSF1–related genes in AML. GeneMANIA was applied to outline HSF1–related functional networks. CancerSEA analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis and Gene Set Enrichment Analysis (GSEA) were performed to mine the potential mechanism of HSF1 in leukemogenesis. Single–sample Gene Set Enrichment Analysis (ssGSEA) was applied to explore the correlation between HSF1 and infiltrating immune cells in AML. Results: HSF1 expression was elevated in AML compared to healthy controls and indicate a poor overall survival. HSF1 expression was significantly correlated with patients age, associated with patient survival in subgroup of bone marrow blasts (%) >20. Functional analyses indicated that HSF1 plays a role in the metastatic status of AML, and is involved in inflammation–related pathways and biological processes. HSF1 expression was significantly correlated with the immune infiltration of nature killer cells and T cell population. Conclusion: HSF1 plays a vital role in the molecular network of AML pathogenesis, and has the potential to be a biomarker for prognosis prediction.
Collapse
Affiliation(s)
- Chunyi Lyu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Qian Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Xuewei Yin
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Zonghong Li
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Teng Wang
- Shandong Key Laboratory of Hematology of Integrated Traditional Chinese and Western Medicine of Health Commission, Institute of Hematology of Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Yan Wang
- Shandong Key Laboratory of Hematology of Integrated Traditional Chinese and Western Medicine of Health Commission, Institute of Hematology of Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China.,Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Siyuan Cui
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Kui Liu
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Zhenzhen Wang
- Shandong Key Laboratory of Hematology of Integrated Traditional Chinese and Western Medicine of Health Commission, Institute of Hematology of Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China.,Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Chang Gao
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Ruirong Xu
- Shandong Key Laboratory of Hematology of Integrated Traditional Chinese and Western Medicine of Health Commission, Institute of Hematology of Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China.,Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| |
Collapse
|
14
|
A novel HSP90 inhibitor SL-145 suppresses metastatic triple-negative breast cancer without triggering the heat shock response. Oncogene 2022; 41:3289-3297. [PMID: 35501463 PMCID: PMC9166677 DOI: 10.1038/s41388-022-02269-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 02/15/2022] [Accepted: 03/03/2022] [Indexed: 12/31/2022]
Abstract
Despite recent advances, there remains a significant unmet need for the development of new targeted therapies for triple-negative breast cancer (TNBC). Although the heat shock protein HSP90 is a promising target, previous inhibitors have had issues during development including undesirable induction of the heat shock response (HSR) and off-target effects leading to toxicity. SL-145 is a novel, rationally-designed C-terminal HSP90 inhibitor that induces apoptosis in TNBC cells via the suppression of oncogenic AKT, MEK/ERK, and JAK2/STAT3 signaling and does not trigger the HSR, in contrast to other inhibitors. In an orthotopic allograft model incorporating breast cancer stem cell-enriched TNBC tumors, SL-145 potently suppressed tumor growth, angiogenesis, and metastases concomitant with dysregulation of the JAK2/STAT3 signaling pathway. Our findings highlight the potential of SL-145 in suppressing metastatic TNBC independent of the HSR.
Collapse
|
15
|
Su J, Dou Z, Hong H, Xu F, Lu X, Lu Q, Ye T, Huang C. KRIBB11: A Promising Drug that Promotes Microglial Process Elongation and Suppresses Neuroinflammation. Front Pharmacol 2022; 13:857081. [PMID: 35370703 PMCID: PMC8971675 DOI: 10.3389/fphar.2022.857081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/03/2022] [Indexed: 11/13/2022] Open
Abstract
Microglia are key components of the central innate immune system. The over-activation of microglia, which occurs in nervous system disorders, is usually accompanied with retractions of their ramified processes. Reversing of microglial process retraction is a potential strategy for the prevention of neuroinflammation. Our previous studies have reported some endogenous molecules and drugs that can promote microglial process elongation at conditions in vitro and in vivo, such as butyrate and β-hydroxybutyrate, sulforaphane, and diallyl disulfide. Here, reported another compound that can promote microglial process elongation. We found that KRIBB11, a compound which has been reported to suppress nitric oxide production in microglia, induced significant elongations of the processes in microglia in cultured and in vivo conditions in a reversible manner. KRIBB11 pretreatment also prevented lipopolysaccharide (LPS)-induced shortenings of microglial process in cultured conditions and in vivo conditions, inflammatory responses in primary cultured microglia and the prefrontal cortex, and depression-like behaviors in mice. Mechanistic studies revealed that KRIBB11 incubation up-regulated phospho-Akt in cultured microglia and Akt inhibition blocked the pro-elongation effect of KRIBB11 on microglial process in cultured conditions and in vivo conditions, suggesting that the regulatory effect of KRIBB11 is Akt-dependent. Akt inhibition was also found to abrogate the preventive effect of KRIBB11 on LPS-induced inflammatory responses in primary cultured microglia and prefrontal cortexes as well as LPS-induced depression-like behaviors in mice. Collectively, our findings demonstrated that KRIBB11 is a novel compound that can prevent microglial activation and neuroinflammation-associated behavioral deficits possibly through inducing the Akt-mediated elongation of microglial process.
Collapse
Affiliation(s)
- Jianbin Su
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, First People’s Hospital of Nantong City, Nantong, China
- *Correspondence: Jianbin Su, ; Chao Huang,
| | - Zhihua Dou
- Department of Pharmacy, Nantong Third Hospital Affiliated to Nantong University, Nantong, China
| | - Hongxiang Hong
- Department of Spine Surgery, Affiliated Hospital 2 of Nantong University, First People’s Hospital of Nantong City, Nantong, China
| | - Feng Xu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, First People’s Hospital of Nantong City, Nantong, China
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| | - Qun Lu
- Department of Pharmacy, Nantong Third Hospital Affiliated to Nantong University, Nantong, China
| | - Ting Ye
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
- *Correspondence: Jianbin Su, ; Chao Huang,
| |
Collapse
|
16
|
Visentin A, Pravato S, Castellani F, Campagnolo M, Angotzi F, Cavarretta CA, Cellini A, Ruocco V, Salvalaggio A, Tedeschi A, Trentin L, Briani C. From Biology to Treatment of Monoclonal Gammopathies of Neurological Significance. Cancers (Basel) 2022; 14:1562. [PMID: 35326711 PMCID: PMC8946535 DOI: 10.3390/cancers14061562] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/06/2022] [Accepted: 03/15/2022] [Indexed: 12/23/2022] Open
Abstract
Monoclonal gammopathy and peripheral neuropathy are common diseases of elderly patients, and almost 10% of patients with neuropathy of unknown cause have paraprotein. However, growing evidence suggests that several hematological malignancies synthesize and release monoclonal proteins that damage the peripheral nervous system through different mechanisms. The spectrum of the disease varies from mild to rapidly progressive symptoms, sometimes affecting not only sensory nerve fibers, but also motor and autonomic fibers. Therefore, a multidisciplinary approach, mainly between hematologists and neurologists, is recommended in order to establish the correct diagnosis of monoclonal gammopathy of neurological significance and to tailor therapy based on specific genetic mutations. In this review, we summarize the spectrum of monoclonal gammopathies of neurological significance, their distinctive clinical and neurophysiological phenotypes, the most relevant pathophysiological events and new therapeutic approaches.
Collapse
Affiliation(s)
- Andrea Visentin
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padova, 35128 Padova, Italy; (S.P.); (F.A.); (C.A.C.); (A.C.); (V.R.); (L.T.)
| | - Stefano Pravato
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padova, 35128 Padova, Italy; (S.P.); (F.A.); (C.A.C.); (A.C.); (V.R.); (L.T.)
| | - Francesca Castellani
- Neurology Unit, Department of Neurosciences, University of Padova, 35128 Padova, Italy; (F.C.); (M.C.); (A.S.); (C.B.)
| | - Marta Campagnolo
- Neurology Unit, Department of Neurosciences, University of Padova, 35128 Padova, Italy; (F.C.); (M.C.); (A.S.); (C.B.)
| | - Francesco Angotzi
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padova, 35128 Padova, Italy; (S.P.); (F.A.); (C.A.C.); (A.C.); (V.R.); (L.T.)
| | - Chiara Adele Cavarretta
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padova, 35128 Padova, Italy; (S.P.); (F.A.); (C.A.C.); (A.C.); (V.R.); (L.T.)
| | - Alessandro Cellini
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padova, 35128 Padova, Italy; (S.P.); (F.A.); (C.A.C.); (A.C.); (V.R.); (L.T.)
| | - Valeria Ruocco
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padova, 35128 Padova, Italy; (S.P.); (F.A.); (C.A.C.); (A.C.); (V.R.); (L.T.)
| | - Alessandro Salvalaggio
- Neurology Unit, Department of Neurosciences, University of Padova, 35128 Padova, Italy; (F.C.); (M.C.); (A.S.); (C.B.)
| | - Alessandra Tedeschi
- ASST Grande Ospedale Metropolitano Niguarda, Niguarda Cancer Center, 20162 Milano, Italy;
| | - Livio Trentin
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padova, 35128 Padova, Italy; (S.P.); (F.A.); (C.A.C.); (A.C.); (V.R.); (L.T.)
| | - Chiara Briani
- Neurology Unit, Department of Neurosciences, University of Padova, 35128 Padova, Italy; (F.C.); (M.C.); (A.S.); (C.B.)
| |
Collapse
|
17
|
Severin F, Urbani A, Varanita T, Bachmann M, Azzolini M, Martini V, Pizzi M, Tos APD, Frezzato F, Mattarei A, Ghia P, Bertilaccio MTS, Gulbins E, Paradisi C, Zoratti M, Semenzato GC, Leanza L, Trentin L, Szabò I. Pharmacological modulation of Kv1.3 potassium channel selectively triggers pathological B lymphocyte apoptosis in vivo in a genetic CLL model. J Exp Clin Cancer Res 2022; 41:64. [PMID: 35172855 PMCID: PMC8848658 DOI: 10.1186/s13046-022-02249-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 01/07/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Ion channels are emerging as promising oncological targets. The potassium channels Kv1.3 and IKCa are highly expressed in the plasma membrane and mitochondria of human chronic lymphocytic leukemia (CLL) cells, compared to healthy lymphocytes. In vitro, inhibition of mitoKv1.3 by PAPTP was shown to kill ex vivo primary human CLL cells, while targeting IKCa with TRAM-34 decreased CLL cell proliferation. METHODS Here we evaluated the effect of the above drugs in CLL cells from ibrutinib-resistant patients and in combination with Venetoclax, two drugs used in the clinical practice. The effects of the drugs were tested also in the Eμ-TCL1 genetic CLL murine model, characterized by a lympho-proliferative disease reminiscent of aggressive human CLL. Eμ-TCL1 mice showing overt disease state were treated with intraperitoneal injections of non-toxic 5 nmol/g PAPTP or 10 nmol/g TRAM-34 once a day and the number and percentage of pathological B cells (CD19+CD5+) in different, pathologically relevant body districts were determined. RESULTS We show that Kv1.3 expression correlates with sensitivity of the human and mouse neoplastic cells to PAPTP. Primary CLL cells from ibrutinib-resistant patients could be killed with PAPTP and this drug enhanced the effect of Venetoclax, by acting on mitoKv1.3 of the inner mitochondrial membrane and triggering rapid mitochondrial changes and cytochrome c release. In vivo, after 2 week- therapy of Eμ-TCL1 mice harboring distinct CLL clones, leukemia burden was reduced by more than 85%: the number and percentage of CLL B cells fall in the spleen and peritoneal cavity and in the peripheral blood, without signs of toxicity. Notably, CLL infiltration into liver and spleen and splenomegaly were also drastically reduced upon PAPTP treatment. In contrast, TRAM-34 did not exert any beneficial effect when administered in vivo to Eμ-TCL1 mice at non-toxic concentration. CONCLUSION Altogether, by comparing vehicle versus compound effect in different Eμ-TCL1 animals bearing unique clones similarly to CLL patients, we conclude that PAPTP significantly reduced leukemia burden in CLL-relevant districts, even in animals with advanced stage of the disease. Our results thus identify PAPTP as a very promising drug for CLL treatment, even for the chemoresistant forms of the disease.
Collapse
Affiliation(s)
- Filippo Severin
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padua School of Medicine, Padua, Italy and Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
| | - Andrea Urbani
- Department of Biomedical Sciences, University of Padua, Padua, Italy.,Department of Biology, University of Padua, Padua, Italy
| | | | | | - Michele Azzolini
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Veronica Martini
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padua School of Medicine, Padua, Italy and Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
| | - Marco Pizzi
- Department of Medicine, Pathology Branch, University of Padua School of Medicine, Padua, Italy
| | - Angelo Paolo Dei Tos
- Department of Medicine, Pathology Branch, University of Padua School of Medicine, Padua, Italy
| | - Federica Frezzato
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padua School of Medicine, Padua, Italy and Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
| | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Paolo Ghia
- Università Vita-Salute San Raffaele and IRCC Ospedale San Raffaele, Milan, Italy
| | | | - Erich Gulbins
- Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany
| | | | - Mario Zoratti
- Department of Biomedical Sciences, University of Padua, Padua, Italy.,CNR Institute of Neurosciences, University of Padua, Padua, Italy
| | - Gianpietro Carlo Semenzato
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padua School of Medicine, Padua, Italy and Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
| | - Luigi Leanza
- Department of Biology, University of Padua, Padua, Italy.
| | - Livio Trentin
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padua School of Medicine, Padua, Italy and Veneto Institute of Molecular Medicine (VIMM), Padua, Italy.
| | - Ildiko Szabò
- Department of Biology, University of Padua, Padua, Italy. .,CNR Institute of Neurosciences, University of Padua, Padua, Italy.
| |
Collapse
|
18
|
Murugan NJ, Voutsadakis IA. Proteasome regulators in pancreatic cancer. World J Gastrointest Oncol 2022; 14:38-54. [PMID: 35116102 PMCID: PMC8790418 DOI: 10.4251/wjgo.v14.i1.38] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/14/2021] [Accepted: 12/02/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic adenocarcinoma is one of the most lethal cancers with rising incidence. Despite progress in its treatment, with the introduction of more effective chemotherapy regimens in the last decade, prognosis of metastatic disease remains inferior to other cancers with long term survival being the exception. Molecular characterization of pancreatic cancer has elucidated the landscape of the disease and has revealed common lesions that contribute to pancreatic carcinogenesis. Regulation of proteostasis is critical in cancers due to increased protein turnover required to support the intense metabolism of cancer cells. The proteasome is an integral part of this regulation and is regulated, in its turn, by key transcription factors, which induce transcription of proteasome structural units. These include FOXO family transcription factors, NFE2L2, hHSF1 and hHSF2, and NF-Y. Networks that encompass proteasome regulators and transduction pathways dysregulated in pancreatic cancer such as the KRAS/ BRAF/MAPK and the Transforming growth factor beta/SMAD pathway contribute to pancreatic cancer progression. This review discusses the proteasome and its transcription factors within the pancreatic cancer cellular micro-environment. We also consider the role of stemness in carcinogenesis and the use of proteasome inhibitors as therapeutic agents.
Collapse
Affiliation(s)
- Nirosha J Murugan
- Department of Biology, Algoma University, Sault Sainte Marie P6A3T6, ON, Canada
| | - Ioannis A Voutsadakis
- Department of Medical Oncology, Sault Area Hospital, Sault Sainte Marie P6A3T6, ON, Canada
| |
Collapse
|
19
|
The Modes of Dysregulation of the Proto-Oncogene T-Cell Leukemia/Lymphoma 1A. Cancers (Basel) 2021; 13:cancers13215455. [PMID: 34771618 PMCID: PMC8582492 DOI: 10.3390/cancers13215455] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 11/19/2022] Open
Abstract
Simple Summary T-cell leukemia/lymphoma 1A (TCL1A) is a proto-oncogene that is mainly expressed in embryonic and fetal tissues, as well as in some lymphatic cells. It is frequently overexpressed in a variety of T- and B-cell lymphomas and in some solid tumors. In chronic lymphocytic leukemia and in T-prolymphocytic leukemia, TCL1A has been implicated in the pathogenesis of these conditions, and high-level TCL1A expression correlates with more aggressive disease characteristics and poorer patient survival. Despite the modes of TCL1A (dys)regulation still being incompletely understood, there are recent advances in understanding its (post)transcriptional regulation. This review summarizes the current concepts of TCL1A’s multi-faceted modes of regulation. Understanding how TCL1A is deregulated and how this can lead to tumor initiation and sustenance can help in future approaches to interfere in its oncogenic actions. Abstract Incomplete biological concepts in lymphoid neoplasms still dictate to a large extent the limited availability of efficient targeted treatments, which entertains the mostly unsatisfactory clinical outcomes. Aberrant expression of the embryonal and lymphatic TCL1 family of oncogenes, i.e., the paradigmatic TCL1A, but also TML1 or MTCP1, is causally implicated in T- and B-lymphocyte transformation. TCL1A also carries prognostic information in these particular T-cell and B-cell tumors. More recently, the TCL1A oncogene has been observed also in epithelial tumors as part of oncofetal stemness signatures. Although the concepts on the modes of TCL1A dysregulation in lymphatic neoplasms and solid tumors are still incomplete, there are recent advances in defining the mechanisms of its (de)regulation. This review presents a comprehensive overview of TCL1A expression in tumors and the current understanding of its (dys)regulation via genomic aberrations, epigenetic modifications, or deregulation of TCL1A-targeting micro RNAs. We also summarize triggers that act through such transcriptional and translational regulation, i.e., altered signals by the tumor microenvironment. A refined mechanistic understanding of these modes of dysregulations together with improved concepts of TCL1A-associated malignant transformation can benefit future approaches to specifically interfere in TCL1A-initiated or -driven tumorigenesis.
Collapse
|
20
|
Targeting of HSP70/HSF1 Axis Abrogates In Vitro Ibrutinib-Resistance in Chronic Lymphocytic Leukemia. Cancers (Basel) 2021; 13:cancers13215453. [PMID: 34771616 PMCID: PMC8582437 DOI: 10.3390/cancers13215453] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/05/2021] [Accepted: 10/27/2021] [Indexed: 12/02/2022] Open
Abstract
Simple Summary The use of ibrutinib has changed the management and clinical history of patients with multiple-treated chronic lymphocytic leukemia (CLL). Nevertheless, an increasing number of patients develop resistance to treatment, with mechanisms still to be fully clarified. Since HSP70 plays a pivotal role in mediating the survival and the progression of CLL, we herein addressed the role of HSP70 and its regulator HSF1 in the development of ibrutinib-mediated resistance. We found an increase in both proteins when the treatment was failing, and thus the disease was progressing. This suggests the involvement of HSP70 in mechanisms of drug resistance. Moreover, we demonstrated that the use, at different levels, of HSP70/HSF1 axis inhibitors could represent a novel rational therapeutic approach to overcome ibrutinib resistance in those patients who relapsed after this type of treatment. Abstract The Btk inhibitor ibrutinib has significantly changed the management of chronic lymphocytic leukemia (CLL) patients. Despite its clinical efficacy, relapses occur, and outcomes after ibrutinib failure are poor. Although BTK and PLCγ2 mutations have been found to be associated with ibrutinib resistance in a fair percentage of CLL patients, no information on resistance mechanisms is available in patients lacking these mutations. The heat shock protein of 70 kDa (HSP70) and its transcription factor heat shock factor 1 (HSF1) play a role in mediating the survival and progression of CLL, as well as taking part in drug resistance in various cancers. We demonstrated that resveratrol and related phenols were able to induce apoptosis in vitro in leukemic cells from CLL untreated patients by acting on the HSP70/HSF1 axis. The same was achieved in cells recovered from 13 CLL patients failing in vivo ibrutinib treatment. HSP70 and HSF1 levels decreased following in vitro treatment, correlating to apoptosis induction. We suggest an involvement of HSP70/HSF1 axis in controlling resistance to ibrutinib in CLL cells, since their inhibition is effective in inducing in vitro apoptosis in cells from ibrutinib refractory patients. The targeting of HSP70/HSF1 axis could represent a novel rational therapeutic strategy for CLL, also for relapsing patients.
Collapse
|
21
|
Proteotoxic Stress as an Exploitable Vulnerability in Cells with Hyperactive AKT. Int J Mol Sci 2021; 22:ijms222111376. [PMID: 34768807 PMCID: PMC8583472 DOI: 10.3390/ijms222111376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 10/05/2021] [Accepted: 10/14/2021] [Indexed: 11/16/2022] Open
Abstract
Hyperactivity of serine-threonine kinase AKT is one of the most common molecular abnormalities in cancer, where it contributes to poor outcomes by facilitating the growth and survival of malignant cells. Despite its well-documented anti-apoptotic effects, hyperactivity of AKT is also known to be stressful to a cell. In an attempt to better elucidate this phenomenon, we observed the signs of proteotoxic stress in cells that harbor hyperactive AKT or have lost its principal negative regulator, PTEN. The activity of HSF1 was predictably elevated under these circumstances. However, such cells proved more sensitive to various regimens of heat shock, including the conditions that were well-tolerated by syngeneic cells without AKT hyperactivity. The sensitizing effect of hyperactive AKT was also seen in HSF1-deficient cells, suggesting that the phenomenon does not require the regulation of HSF1 by this kinase. Notably, the elevated activity of AKT was accompanied by increased levels of XBP1, a key component of cell defense against proteotoxic stress. Interestingly, the cells harboring hyperactive AKT were also more dependent on XBP1 for their growth. Our observations suggest that proteotoxic stress conferred by hyperactive AKT represents a targetable vulnerability, which can be exploited by either elevating the stress above the level tolerated by such cells or by eliminating the factors that enable such tolerance.
Collapse
|
22
|
Edwards SL, Erdenebat P, Morphis AC, Kumar L, Wang L, Chamera T, Georgescu C, Wren JD, Li J. Insulin/IGF-1 signaling and heat stress differentially regulate HSF1 activities in germline development. Cell Rep 2021; 36:109623. [PMID: 34469721 PMCID: PMC8442575 DOI: 10.1016/j.celrep.2021.109623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 05/25/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022] Open
Abstract
Germline development is sensitive to nutrient availability and environmental perturbation. Heat shock transcription factor 1 (HSF1), a key transcription factor driving the cellular heat shock response (HSR), is also involved in gametogenesis. The precise function of HSF1 (HSF-1 in C. elegans) and its regulation in germline development are poorly understood. Using the auxin-inducible degron system in C. elegans, we uncovered a role of HSF-1 in progenitor cell proliferation and early meiosis and identified a compact but important transcriptional program of HSF-1 in germline development. Interestingly, heat stress only induces the canonical HSR in a subset of germ cells but impairs HSF-1 binding at its developmental targets. Conversely, insulin/insulin growth factor 1 (IGF-1) signaling dictates the requirement for HSF-1 in germline development and functions through repressing FOXO/DAF-16 in the soma to activate HSF-1 in germ cells. We propose that this non-cell-autonomous mechanism couples nutrient-sensing insulin/IGF-1 signaling to HSF-1 activation to support homeostasis in rapid germline growth.
Collapse
Affiliation(s)
- Stacey L Edwards
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Purevsuren Erdenebat
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Allison C Morphis
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Lalit Kumar
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Lai Wang
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Tomasz Chamera
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Constantin Georgescu
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Jonathan D Wren
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Jian Li
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
| |
Collapse
|
23
|
Zhao N, Zhang J, Zhao L, Fu X, Zhao Q, Chao M, Cao H, Jiao Y, Hu Y, Chen C, Wang L, Wang H. Long Noncoding RNA NONHSAT079852.2 Contributes to GBM Recurrence by Functioning as a ceRNA for has-mir-10401-3p to Facilitate HSPA1A Upregulation. Front Oncol 2021; 11:636632. [PMID: 34307121 PMCID: PMC8297974 DOI: 10.3389/fonc.2021.636632] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/22/2021] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common brain malignancy and major cause of high mortality in patients with GBM, and its high recurrence rate is its most prominent feature. However, the pathobiological mechanisms involved in recurrent GBM remain largely unknown. Here, whole-transcriptome sequencing (RNA-sequencing, RNA-Seq) was used in characterizing the expression profile of recurrent GBM, and the aim was to identify crucial biomarkers that contribute to GBM relapse. Differentially expressed RNAs in three recurrent GBM tissues compared with three primary GBM tissues were identified through RNA-Seq. The function and mechanism of a candidate long noncoding RNA (lncRNA) in the progression and recurrence of GBM were elucidated by performing comprehensive bioinformatics analyses, such as functional enrichment analysis, protein-protein interaction prediction, and lncRNA-miRNA-mRNA regulatory network construction, and a series of in vitro assays. As the most significantly upregulated gene identified in recurrent GBM, HSPA1A is mainly related to antigen presentation and the MAPK signaling pathway, as indicated by functional enrichment analysis. HSPA1A was predicted as the target gene of the lncRNA NONHSAT079852.2. qRT-PCR revealed that NONHSAT079852.2 was significantly elevated in recurrent GBM relative to that in primary GBM, and high NONHSAT079852.2 expression was associated with the poor overall survival rates of patients with GBM. The knockdown of NONHSAT079852.2 successfully induced tumor cell apoptosis, inhibited the proliferation, migration, invasion and the expression level of HSPA1A in glioma cells. NONHSAT079852.2 was identified to be a sponge for hsa-miR-10401-3p through luciferase reporter assay. Moreover, HSPA1A was targeted and regulated by hsa-miR-10401-3p. Collectively, the results suggested that NONHSAT079852.2 acts as a sponge of hsa-mir-10401-3p and thereby enhances HSPA1A expression, promotes tumor cell proliferation and invasion, and leads to the progression and recurrence of GBM. This study will provide new insight into the regulatory mechanisms of NONHSAT079852.2-mediated competing endogenous RNA in the pathogenesis of recurrent GBM and evidence of the potential of lncRNAs as diagnostic biomarkers or potential therapeutic targets.
Collapse
Affiliation(s)
- Ningning Zhao
- College of Life Sciences, Northwest University, Xian, China
| | - Jiajie Zhang
- College of Life Sciences, Northwest University, Xian, China
| | - Lili Zhao
- College of Life Sciences, Northwest University, Xian, China
| | - Xiaoni Fu
- College of Life Sciences, Northwest University, Xian, China
| | - Qian Zhao
- College of Life Sciences, Northwest University, Xian, China
| | - Min Chao
- Department of Neurosurgery, Tangdu Hospital of Air Force Medical University, Xian, China
| | - Haiyan Cao
- Department of Neurosurgery, Tangdu Hospital of Air Force Medical University, Xian, China
| | - Yang Jiao
- Department of Neurosurgery, Tangdu Hospital of Air Force Medical University, Xian, China
| | - Yaqin Hu
- Department of Neurosurgery, Tangdu Hospital of Air Force Medical University, Xian, China
| | - Chao Chen
- College of Life Sciences, Northwest University, Xian, China
| | - Liang Wang
- Department of Neurosurgery, Tangdu Hospital of Air Force Medical University, Xian, China
| | - Huijuan Wang
- College of Life Sciences, Northwest University, Xian, China
| |
Collapse
|
24
|
In vitro maturation in the presence of Leukemia Inhibitory Factor modulates gene and miRNA expression in bovine oocytes and embryos. Sci Rep 2020; 10:17777. [PMID: 33082423 PMCID: PMC7575586 DOI: 10.1038/s41598-020-74961-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023] Open
Abstract
Members of the interleukin-6 (IL-6) family of cytokines are important for reproductive function that are mediated through changes in gene and miRNA expression. Herein, we characterized the expression of miR-21, miR-155, miR-34c and miR-146a in bovine oocytes and cumulus cells during in vitro maturation (IVM) with leukemia inhibitory factor (LIF), IL-6 and IL-11 or unsupplemented controls. LIF-exposed COCs showed higher expression of miR-21 and miR-155 in oocytes, whereas miR-146a expression was increased in oocytes matured with IL-6 and IL-11. In cumulus cells, miR-155 expression was elevated by all treatments while only LIF increased miR-21 expression. Based on these results, we next examined how LIF exposure during IVM affected oocyte competence, through IVF and the expression of specific genes in GV- and MII-oocytes, in 2- and 8-cell embryos, and in Day 8-blastocysts. LIF supplementation did not affect cleavage rate, blastocyst yield or several other developmental parameters, but did increase hatching rate. LIF suppressed DPPA3, ZAR1 and NPM2 expression in 2 cell- and/or 8-cell embryos. LIF increased the expression of KAT2A and HSPA1A in MII-oocytes, and that of HDAC1, KAT2A and HSP90AA1 and the BAX:BCL2L1 ratio in 2-cell embryos. In contrast, HDAC1, KAT2A and HSP90AA1 expression and BAX:BCL2L1 ratio was lower in 8-cell embryos derived from LIF oocytes. IVM with LIF also increased the expression of DNMT3A, HSPA1A and HSP90AA1 in blastocysts. In conclusion, supplementation with LIF during IVM was consistently associated with changes in the relative abundance of transcripts in mature bovine oocytes and in specific embryo developmental stages.
Collapse
|
25
|
Visentin A, Frezzato F, Severin F, Imbergamo S, Pravato S, Romano Gargarella L, Manni S, Pizzo S, Ruggieri E, Facco M, Brunati AM, Semenzato G, Piazza F, Trentin L. Lights and Shade of Next-Generation Pi3k Inhibitors in Chronic Lymphocytic Leukemia. Onco Targets Ther 2020; 13:9679-9688. [PMID: 33061448 PMCID: PMC7532889 DOI: 10.2147/ott.s268899] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022] Open
Abstract
The treatment (i.e. therapy and management) of chronic lymphocytic leukemia (i.e. the disease) has been improved thanks to the introduction (i.e. approval) of kinase inhibitors during the last years. PI3K is one of the most important kinases at the crossroad to the B-cell receptor and cytokine receptor which play a key role in CLL cell survival, proliferation and migration. Idelalisib is the first in class PI3Kδ inhibitor approved for the treatment of relapsed/refractory CLL in combination with rituximab. Idelalisib activity in heavily treated patients is balanced by recurrent adverse events which limit its long-term use. These limitations prompt the investigation on novel PI3K inhibitors, also targeting different protein isoforms, and alternative schedule strategies. In this regard, duvelisib is the only PI3K γ and δ inhibitor approved as single agent for relapsed CLL. In this review, we will address novel insights on PI3K structure, isoforms, regulating signaling and the most updated data of next-generation PI3K inhibitors in CLL.
Collapse
Affiliation(s)
- Andrea Visentin
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| | - Federica Frezzato
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| | - Filippo Severin
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| | - Silvia Imbergamo
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Stefano Pravato
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Leila Romano Gargarella
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Sabrina Manni
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| | - Serena Pizzo
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| | - Edoardo Ruggieri
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| | - Monica Facco
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| | | | - Gianpietro Semenzato
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| | - Francesco Piazza
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| | - Livio Trentin
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| |
Collapse
|
26
|
Briani C, Visentin A, Cerri F, Quattrini A. From pathogenesis to personalized treatments of neuropathies in hematological malignancies. J Peripher Nerv Syst 2020; 25:212-221. [PMID: 32686258 DOI: 10.1111/jns.12405] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 12/21/2022]
Abstract
The peripheral nervous system may be involved at any stage in the course of several hematological diseases, the most common being monoclonal gammopathies (of undetermined significance or malignant) or lymphomas. The underlying pathogenic mechanisms are different and therapies aim at targeting the dangerous either B-cell or plasma cell clones. Recently, high-throughput technologies, and next-generation sequencing have increased our knowledge of hematological diseases pathogenesis by the identification of somatic mutation affecting pivotal signaling pathways. Accordingly, new target therapies are used that may also be borrowed for treatment of neuropathies in hematological diseases.
Collapse
Affiliation(s)
- Chiara Briani
- Department of Neuroscience, University of Padova, Padova, Italy
| | - Andrea Visentin
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padova, Padova, Italy
| | - Federica Cerri
- Experimental Neuropathology Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Angelo Quattrini
- Experimental Neuropathology Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
27
|
Xia C, Liu C, He Z, Cai Y, Chen J. Metformin inhibits cervical cancer cell proliferation by modulating PI3K/Akt-induced major histocompatibility complex class I-related chain A gene expression. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:127. [PMID: 32631421 PMCID: PMC7336474 DOI: 10.1186/s13046-020-01627-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 06/21/2020] [Indexed: 02/07/2023]
Abstract
Background Recent studies have shown that the classic hypoglycemic drug metformin inhibits tumor growth; however, the underlying mechanism remains unclear. We previously showed that metformin disrupts the sponge effect of long non-coding RNA MALAT1/miR-142-3p to inhibit cervical cancer cell proliferation. In this study, we interrogated the ability of metformin to modulate the anti-tumor immune response in cervical cancer. Methods The cell counting kit-8 assay was used to detect the viability of cervical cancer cells. Flow cytometry assays were performed to measure cell apoptosis and cell cycle. Lactate dehydrogenase (LDH) cytotoxicity assay was used to detect NK Cell Cytotoxicity. Relative protein levels were determined by immunoblotting and relative gene levels were determined by quantitative real-time PCR. Tumor Xenograft Modeling was used to evaluate the effect of metformin in vivo. Results Metformin inhibited cervical cancer cell proliferation, cervical cancer xenograft growth, expression of PCNA, p-PI3K and p-Akt. Moreover metformin induced cervical cancer cell apoptosis and caused cancer cell cycle arrest. In addition, metformin upregulated the expression of DDR-1 and p53 in human cervical cancer cells. Furthermore, metformin also regulated the mRNA and protein expression of MICA and HSP70 on the surface of human cervical cancer cells via the PI3K/Akt pathway, enhancing NK cell cytotoxicity. Conclusions In conclusion, our results suggest that metformin may be used as immunopotentiator to inhibit cervical cancer progression and may be considered a viable candidate for combination therapy with immunotherapy.
Collapse
Affiliation(s)
- Chenglai Xia
- South Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, 528000, China. .,School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510150, China.
| | - Chang Liu
- South Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, 528000, China.,Foshan Women and Child hospital, Foshan, 528000, China
| | - Zhihong He
- South Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, 528000, China.,Foshan Women and Child hospital, Foshan, 528000, China
| | - Yantao Cai
- South Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, 528000, China.,Foshan Women and Child hospital, Foshan, 528000, China
| | - Jinman Chen
- South Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, 528000, China.,Foshan Women and Child hospital, Foshan, 528000, China
| |
Collapse
|
28
|
Specific Roles of HSP27 S15 Phosphorylation Augmenting the Nuclear Function of HER2 to Promote Trastuzumab Resistance. Cancers (Basel) 2020; 12:cancers12061540. [PMID: 32545363 PMCID: PMC7352409 DOI: 10.3390/cancers12061540] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/09/2020] [Accepted: 06/09/2020] [Indexed: 02/07/2023] Open
Abstract
Trastuzumab (TZMB) is widely used as first line therapy for breast cancer (BC) patients overexpressing human epidermal growth factor receptor 2 (HER2). Despite its clinical benefits, many patients suffer from primary or secondary resistance to this drug within one year. As diverse molecular mechanisms occur contemporaneously during the resistance development, we focused on elucidating the role of heat shock protein 27 (HSP27) in TZMB-resistance, as this protein simultaneously regulates the function of diverse client molecules that are involved in the resistance mechanism. By extensively utilizing TZMB-refractory breast cancer cell lines transduced with diverse phosphovariants of HSP27, our study newly revealed that specific phosphorylation of HSP27 at S15 promoted its S78 phosphorylation and served as key mediator to promote direct interactions that increase the stability of HER2 and protein kinase B (AKT). This phosphorylation promoted nuclear translocation of HER2, enhancing the distinct nuclear function of HER2 that promoted AKT activation and cyclin D1 expression. Co-administration of TZMB and a functional inhibitor of HSP27, J2, significantly reduced the S15/78 phosphorylation of HSP27, which downregulated HER2 and its downstream signals, sensitizing TZMB-refractory cell, and JIMT1-xenograft mouse models to TZMB. Collectively, p-HSP27S15 could serve as a valuable predictive marker and also a therapeutic target for TZMB-resistance.
Collapse
|
29
|
Alasady MJ, Mendillo ML. The Multifaceted Role of HSF1 in Tumorigenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1243:69-85. [PMID: 32297212 DOI: 10.1007/978-3-030-40204-4_5] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Heat Shock Factor 1 (HSF1), the master transcriptional regulator of the heat shock response (HSR), was first cloned more than 30 years ago. Most early research interrogating the role that HSF1 plays in biology focused on its cytoprotective functions, as a factor that promotes the survival of organisms by protecting against the proteotoxicity associated with neurodegeneration and other pathological conditions. However, recent studies have revealed a deleterious role of HSF1, as a factor that is co-opted by cancer cells to promote their own survival to the detriment of the organism. In cancer, HSF1 operates in a multifaceted manner to promote oncogenic transformation, proliferation, metastatic dissemination, and anti-cancer drug resistance. Here we review our current understanding of HSF1 activation and function in malignant progression and discuss the potential for HSF1 inhibition as a novel anticancer strategy. Collectively, this ever-growing body of work points to a prominent role of HSF1 in nearly every aspect of carcinogenesis.
Collapse
Affiliation(s)
- Milad J Alasady
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Simpson Querrey Center for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Marc L Mendillo
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA. .,Simpson Querrey Center for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA. .,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
30
|
Gao D, Zhou Z, Huang H. miR-30b-3p Inhibits Proliferation and Invasion of Hepatocellular Carcinoma Cells via Suppressing PI3K/Akt Pathway. Front Genet 2019; 10:1274. [PMID: 31921311 PMCID: PMC6923265 DOI: 10.3389/fgene.2019.01274] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/19/2019] [Indexed: 12/24/2022] Open
Abstract
Background: The micro-RNA miR-30b-3p has been reported to play a crucial role in several cancers. However, the biological function of miR-30b-3p in hepatocellular carcinoma (HCC) is still unknown. Methods: RT-qPCR was employed to determine the expression of miR-30b-3p in HCC tissues and cells. The MTT assay, colony formation assay, and cell migration and invasion assay were employed to evaluate the role of miR-30b-3p in HCC cells. A dual-luciferase reporter assay was employed to verify the target of miR-30b-3p. Western blotting was employed to determine the expression of key molecular signal transducers along TRIM27-PI3K/Akt axis. Results: Expression of miR-30b-3p was markedly decreased in HCC tissues and cells and positively correlated with higher overall survival. Moreover, miR-30b-3p overexpression significantly repressed cell viability, proliferation, migration, and invasion of HCC cells in vitro. Notably, we demonstrated that miR-30b-3p directly bound to the 3′-untranslated region of tripartite motif containing 27 (TRIM27) mRNA by downregulating the expression of TRIM27, which was demonstrated to be negatively correlated with miR-30b-3p expression. TRIM27 was demonstrated to have an oncogenic role in HCC cells by enhancing cell viability, proliferation, migration, and invasion. Finally, the miR-30-3p-TRIM27-PI3K/Akt axis was shown to play a crucial role in HCC cells in vitro. Conclusion: Our results indicated that miR-30-3p might act as a new biomarker for the future diagnosis and treatment HCC.
Collapse
Affiliation(s)
- Dongkai Gao
- Department of Infectious Diseases, Zhuji People's Hospital of Zhejiang Province, Shaoxing, China
| | - Zumo Zhou
- Department of Infectious Diseases, Zhuji People's Hospital of Zhejiang Province, Shaoxing, China
| | - Heqing Huang
- Department of Infectious Diseases, Zhuji People's Hospital of Zhejiang Province, Shaoxing, China
| |
Collapse
|
31
|
In Chronic Lymphocytic Leukemia the JAK2/STAT3 Pathway Is Constitutively Activated and Its Inhibition Leads to CLL Cell Death Unaffected by the Protective Bone Marrow Microenvironment. Cancers (Basel) 2019; 11:cancers11121939. [PMID: 31817171 PMCID: PMC6966457 DOI: 10.3390/cancers11121939] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/11/2019] [Accepted: 11/29/2019] [Indexed: 01/01/2023] Open
Abstract
The bone marrow microenvironment promotes proliferation and drug resistance in chronic lymphocytic leukemia (CLL). Although ibrutinib is active in CLL, it is rarely able to clear leukemic cells protected by bone marrow mesenchymal stromal cells (BMSCs) within the marrow niche. We investigated the modulation of JAK2/STAT3 pathway in CLL by BMSCs and its targeting with AG490 (JAK2 inhibitor) or Stattic (STAT3 inhibitor). B cells collected from controls and CLL patients, were treated with medium alone, ibrutinib, JAK/Signal Transducer and Activator of Transcription (STAT) inhibitors, or both drugs, in the presence of absence of BMSCs. JAK2/STAT3 axis was evaluated by western blotting, flow cytometry, and confocal microscopy. We demonstrated that STAT3 was phosphorylated in Tyr705 in the majority of CLL patients at basal condition, and increased following co-cultures with BMSCs or IL-6. Treatment with AG490, but not Stattic, caused STAT3 and Lyn dephosphorylation, through re-activation of SHP-1, and triggered CLL apoptosis even when leukemic cells were cultured on BMSC layers. Moreover, while BMSCs hamper ibrutinib activity, the combination of ibrutinib+JAK/STAT inhibitors increase ibrutinib-mediated leukemic cell death, bypassing the pro-survival stimuli derived from BMSCs. We herein provide evidence that JAK2/STAT3 signaling might play a key role in the regulation of CLL-BMSC interactions and its inhibition enhances ibrutinib, counteracting the bone marrow niche.
Collapse
|