1
|
Kwantwi LB, Rosen ST, Querfeld C. The Tumor Microenvironment as a Therapeutic Target in Cutaneous T Cell Lymphoma. Cancers (Basel) 2024; 16:3368. [PMID: 39409988 PMCID: PMC11482616 DOI: 10.3390/cancers16193368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 09/27/2024] [Accepted: 09/28/2024] [Indexed: 10/19/2024] Open
Abstract
Cutaneous T cell lymphomas (CTCLs) are a heterogeneous group of non-Hodgkin lymphomas, with mycosis fungoides and Sézary syndrome being the two common subtypes. Despite the substantial improvement in early-stage diagnosis and treatments, some patients still progress to the advanced stage with an elusive underpinning mechanism. While this unsubstantiated disease mechanism coupled with diverse clinical outcomes poses challenges in disease management, emerging evidence has implicated the tumor microenvironment in the disease process, thus revealing a promising therapeutic potential of targeting the tumor microenvironment. Notably, malignant T cells can shape their microenvironment to dampen antitumor immunity, leading to Th2-dominated responses that promote tumor progression. This is largely orchestrated by alterations in cytokines expression patterns, genetic dysregulations, inhibitory effects of immune checkpoint molecules, and immunosuppressive cells. Herein, the recent insights into the determining factors in the CTCL tumor microenvironment that support their progression have been highlighted. Also, recent advances in strategies to target the CTCL tumor micromovement with the rationale of improving treatment efficacy have been discussed.
Collapse
Affiliation(s)
- Louis Boafo Kwantwi
- Department of Pathology, City of Hope Medical Center, Duarte, CA 91010, USA
- Beckman Research Institute, Duarte, CA 91010, USA
- Department of Anatomy and Neurobiology, College of Medicine, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Steven T Rosen
- Beckman Research Institute, Duarte, CA 91010, USA
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, CA 91010, USA
| | - Christiane Querfeld
- Department of Pathology, City of Hope Medical Center, Duarte, CA 91010, USA
- Beckman Research Institute, Duarte, CA 91010, USA
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, CA 91010, USA
- Division of Dermatology, City of Hope Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
2
|
Guan M, Zhao H, Zhang Q, Li L, Wang X, Tang B. A novel anoikis-related signature predicts prognosis risk and treatment responsiveness in diffuse large B-cell lymphoma. Expert Rev Mol Diagn 2024; 24:439-457. [PMID: 38709202 DOI: 10.1080/14737159.2024.2351465] [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: 06/02/2023] [Accepted: 03/05/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND Although anoikis plays a role in cancer metastasis and aggressiveness, it has rarely been reported in diffuse large B cell lymphoma (DLBCL). METHODS We obtained RNA sequencing data and matched clinical data from the GEO database. An anoikis-related genes (ARGs)-based risk signature was developed in GSE10846 training cohort and validated in three other cohorts. Additionally, we predicted half-maximal inhibitory concentration (IC50) of drugs based on bioinformatics method and obtained the actual IC50 to some chemotherapy drugs via cytotoxicity assay. RESULTS The high-risk group, as determined by our signature, was associated with worse prognosis and an immunosuppressive environment in DLBCL. Meanwhile, the nomogram based on eight variables had more accurate ability in forecasting the prognosis than the international prognostic index in DLBCL. The prediction of IC50 indicated that DLBCL patients in the high-risk group were more sensitive to doxorubicin, IPA-3, lenalidomide, gemcitabine, and CEP.701, while patients in the low-risk group were sensitive to cisplatin and dasatinib. Consistent with the prediction, cytotoxicity assay suggested the higher sensitivity to doxorubicin and gemcitabine and the lower sensitivity to dasatinib in the high-risk group in DLBCL. CONCLUSION The ARG-based signature may provide a promising direction for prognosis prediction and treatment optimization for DLBCL patients.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/diagnosis
- Prognosis
- Anoikis/drug effects
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Biomarkers, Tumor/genetics
- Transcriptome
- Antineoplastic Agents/therapeutic use
- Antineoplastic Agents/pharmacology
- Nomograms
Collapse
Affiliation(s)
- Mingze Guan
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Hua Zhao
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Qi Zhang
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Li Li
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Xiaobo Wang
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Bo Tang
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| |
Collapse
|
3
|
Luo Y, de Gruijl FR, Vermeer MH, Tensen CP. "Next top" mouse models advancing CTCL research. Front Cell Dev Biol 2024; 12:1372881. [PMID: 38665428 PMCID: PMC11044687 DOI: 10.3389/fcell.2024.1372881] [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: 01/18/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
This review systematically describes the application of in vivo mouse models in studying cutaneous T-cell lymphoma (CTCL), a complex hematological neoplasm. It highlights the diverse research approaches essential for understanding CTCL's intricate pathogenesis and evaluating potential treatments. The review categorizes various mouse models, including xenograft, syngeneic transplantation, and genetically engineered mouse models (GEMMs), emphasizing their contributions to understanding tumor-host interactions, gene functions, and studies on drug efficacy in CTCL. It acknowledges the limitations of these models, particularly in fully replicating human immune responses and early stages of CTCL. The review also highlights novel developments focusing on the potential of skin-targeted GEMMs in studying natural skin lymphoma progression and interactions with the immune system from onset. In conclusion, a balanced understanding of these models' strengths and weaknesses are essential for accelerating the deciphering of CTCL pathogenesis and developing treatment methods. The GEMMs engineered to target specifically skin-homing CD4+ T cells can be the next top mouse models that pave the way for exploring the effects of CTCL-related genes.
Collapse
Affiliation(s)
| | | | | | - Cornelis P. Tensen
- Department of Dermatology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
4
|
Sorger H, Dey S, Vieyra‐Garcia PA, Pölöske D, Teufelberger AR, de Araujo ED, Sedighi A, Graf R, Spiegl B, Lazzeri I, Braun T, Garces de los Fayos Alonso I, Schlederer M, Timelthaler G, Kodajova P, Pirker C, Surbek M, Machtinger M, Graier T, Perchthaler I, Pan Y, Fink‐Puches R, Cerroni L, Ober J, Otte M, Albrecht JD, Tin G, Abdeldayem A, Manaswiyoungkul P, Olaoye OO, Metzelder ML, Orlova A, Berger W, Wobser M, Nicolay JP, André F, Nguyen VA, Neubauer HA, Fleck R, Merkel O, Herling M, Heitzer E, Gunning PT, Kenner L, Moriggl R, Wolf P. Blocking STAT3/5 through direct or upstream kinase targeting in leukemic cutaneous T-cell lymphoma. EMBO Mol Med 2022; 14:e15200. [PMID: 36341492 PMCID: PMC9727928 DOI: 10.15252/emmm.202115200] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 09/26/2022] [Accepted: 10/02/2022] [Indexed: 11/09/2022] Open
Abstract
Leukemic cutaneous T-cell lymphomas (L-CTCL) are lymphoproliferative disorders of skin-homing mature T-cells causing severe symptoms and high mortality through chronic inflammation, tissue destruction, and serious infections. Despite numerous genomic sequencing efforts, recurrent driver mutations have not been identified, but chromosomal losses and gains are frequent and dominant. We integrated genomic landscape analyses with innovative pharmacologic interference studies to identify key vulnerable nodes in L-CTCL. We detected copy number gains of loci containing the STAT3/5 oncogenes in 74% (n = 17/23) of L-CTCL, which correlated with the increased clonal T-cell count in the blood. Dual inhibition of STAT3/5 using small-molecule degraders and multi-kinase blockers abolished L-CTCL cell growth in vitro and ex vivo, whereby PAK kinase inhibition was specifically selective for L-CTCL patient cells carrying STAT3/5 gains. Importantly, the PAK inhibitor FRAx597 demonstrated encouraging anti-leukemic activity in vivo by inhibiting tumor growth and disease dissemination in intradermally xenografted mice. We conclude that STAT3/5 and PAK kinase interaction represents a new therapeutic node to be further explored in L-CTCL.
Collapse
Affiliation(s)
- Helena Sorger
- Unit of Functional Cancer Genomics, Institute of Animal Breeding and GeneticsUniversity of Veterinary MedicineViennaAustria
- Department of Pediatric and Adolescent Surgery, Vienna General HospitalMedical University of ViennaViennaAustria
| | - Saptaswa Dey
- Department of Dermatology and VenereologyMedical University of GrazGrazAustria
- Department of PathologyMedical University of ViennaViennaAustria
| | | | - Daniel Pölöske
- Unit of Functional Cancer Genomics, Institute of Animal Breeding and GeneticsUniversity of Veterinary MedicineViennaAustria
| | | | - Elvin D de Araujo
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaONCanada
- Centre for Medicinal ChemistryUniversity of Toronto MississaugaMississaugaONCanada
| | - Abootaleb Sedighi
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaONCanada
- Centre for Medicinal ChemistryUniversity of Toronto MississaugaMississaugaONCanada
| | - Ricarda Graf
- Diagnostic & Research Center for Molecular Bio‐Medicine, Institute of Human GeneticsMedical University of GrazGrazAustria
| | - Benjamin Spiegl
- Diagnostic & Research Center for Molecular Bio‐Medicine, Institute of Human GeneticsMedical University of GrazGrazAustria
| | - Isaac Lazzeri
- Diagnostic & Research Center for Molecular Bio‐Medicine, Institute of Human GeneticsMedical University of GrazGrazAustria
| | - Till Braun
- Department of Medicine ICIO‐ABCD, CECAD and CMMC Cologne UniversityCologneGermany
| | - Ines Garces de los Fayos Alonso
- Department of PathologyMedical University of ViennaViennaAustria
- Unit of Laboratory Animal PathologyUniversity of Veterinary Medicine ViennaViennaAustria
| | | | | | - Petra Kodajova
- Unit of Laboratory Animal PathologyUniversity of Veterinary Medicine ViennaViennaAustria
| | - Christine Pirker
- Centre for Cancer ResearchMedical University of ViennaViennaAustria
- Comprehensive Cancer CenterMedical University of ViennaViennaAustria
| | - Marta Surbek
- Unit of Functional Cancer Genomics, Institute of Animal Breeding and GeneticsUniversity of Veterinary MedicineViennaAustria
| | - Michael Machtinger
- Unit of Functional Cancer Genomics, Institute of Animal Breeding and GeneticsUniversity of Veterinary MedicineViennaAustria
| | - Thomas Graier
- Department of Dermatology and VenereologyMedical University of GrazGrazAustria
| | | | - Yi Pan
- Department of Dermatology and VenereologyMedical University of GrazGrazAustria
| | - Regina Fink‐Puches
- Department of Dermatology and VenereologyMedical University of GrazGrazAustria
| | - Lorenzo Cerroni
- Department of Dermatology and VenereologyMedical University of GrazGrazAustria
| | - Jennifer Ober
- Core Facility Flow Cytometry, Center for Medical Research (ZMF)Medical University of GrazGrazAustria
| | - Moritz Otte
- Department of Medicine ICIO‐ABCD, CECAD and CMMC Cologne UniversityCologneGermany
| | - Jana D Albrecht
- Department of DermatologyUniversity Hospital MannheimMannheimGermany
| | - Gary Tin
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaONCanada
- Centre for Medicinal ChemistryUniversity of Toronto MississaugaMississaugaONCanada
| | - Ayah Abdeldayem
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaONCanada
- Centre for Medicinal ChemistryUniversity of Toronto MississaugaMississaugaONCanada
| | - Pimyupa Manaswiyoungkul
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaONCanada
- Centre for Medicinal ChemistryUniversity of Toronto MississaugaMississaugaONCanada
| | - Olasunkanmi O Olaoye
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaONCanada
- Centre for Medicinal ChemistryUniversity of Toronto MississaugaMississaugaONCanada
| | - Martin L Metzelder
- Department of Pediatric and Adolescent Surgery, Vienna General HospitalMedical University of ViennaViennaAustria
| | - Anna Orlova
- Unit of Functional Cancer Genomics, Institute of Animal Breeding and GeneticsUniversity of Veterinary MedicineViennaAustria
| | - Walter Berger
- Centre for Cancer ResearchMedical University of ViennaViennaAustria
- Comprehensive Cancer CenterMedical University of ViennaViennaAustria
| | - Marion Wobser
- Department of DermatologyUniversity Hospital WuerzburgWuerzburgGermany
| | - Jan P Nicolay
- Department of DermatologyUniversity Hospital MannheimMannheimGermany
| | - Fiona André
- University Clinic for Dermatology, Venereology and Allergology InnsbruckMedical University of InnsbruckInnsbruckAustria
| | - Van Anh Nguyen
- University Clinic for Dermatology, Venereology and Allergology InnsbruckMedical University of InnsbruckInnsbruckAustria
| | - Heidi A Neubauer
- Unit of Functional Cancer Genomics, Institute of Animal Breeding and GeneticsUniversity of Veterinary MedicineViennaAustria
| | | | - Olaf Merkel
- Department of PathologyMedical University of ViennaViennaAustria
| | - Marco Herling
- Department of Medicine ICIO‐ABCD, CECAD and CMMC Cologne UniversityCologneGermany
- Department of Hematology, Cellular Therapy, and HemostaseologyUniversity of LeipzigLeipzigGermany
| | - Ellen Heitzer
- Diagnostic & Research Center for Molecular Bio‐Medicine, Institute of Human GeneticsMedical University of GrazGrazAustria
| | - Patrick T Gunning
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaONCanada
- Centre for Medicinal ChemistryUniversity of Toronto MississaugaMississaugaONCanada
- Janpix, a Centessa CompanyLondonUK
| | - Lukas Kenner
- Department of PathologyMedical University of ViennaViennaAustria
- Unit of Laboratory Animal PathologyUniversity of Veterinary Medicine ViennaViennaAustria
- Comprehensive Cancer CenterMedical University of ViennaViennaAustria
- Christian Doppler Laboratory for Applied Metabolomics (CDL‐AM), Division of Nuclear MedicineMedical University of ViennaViennaAustria
- CBmed GmbH Center for Biomarker Research in MedicineGrazAustria
| | - Richard Moriggl
- Unit of Functional Cancer Genomics, Institute of Animal Breeding and GeneticsUniversity of Veterinary MedicineViennaAustria
| | - Peter Wolf
- Department of Dermatology and VenereologyMedical University of GrazGrazAustria
- BioTechMed GrazGrazAustria
| |
Collapse
|
5
|
p21-Activated kinases as promising therapeutic targets in hematological malignancies. Leukemia 2022; 36:315-326. [PMID: 34697424 DOI: 10.1038/s41375-021-01451-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/30/2021] [Accepted: 10/07/2021] [Indexed: 01/12/2023]
Abstract
The p21-Activated Kinases (PAKs) are a family of six serine/threonine kinases that were originally identified as downstream effectors of the Rho GTPases Cdc42 and Rac. Since the first PAK was discovered in 1994, studies have revealed their fundamental and biological importance in the development of physiological systems. Within the cell, PAKs also play significant roles in regulating essential cellular processes such as cytoskeletal dynamics, gene expression, cell survival, and cell cycle progression. These processes are often deregulated in numerous cancers when different PAKs are overexpressed or amplified at the chromosomal level. Furthermore, PAKs modulate multiple oncogenic signaling pathways which facilitate apoptosis escape, uncontrolled proliferation, and drug resistance. There is growing insight into the critical roles of PAKs in regulating steady-state hematopoiesis, including the properties of hematopoietic stem cells (HSC), and the initiation and progression of hematological malignancies. This review will focus on the most recent studies that provide experimental evidence showing how specific PAKs regulate the properties of leukemic stem cells (LSCs) and drug-resistant cells to initiate and maintain hematological malignancies. The current understanding of the molecular and cellular mechanisms by which the PAKs operate in specific human leukemia or lymphomas will be discussed. From a translational point of view, PAKs have been suggested to be critical therapeutic targets and potential prognosis markers; thus, this review will also discuss current therapeutic strategies against hematological malignancies using existing small-molecule PAK inhibitors, as well as promising combination treatments, to sensitize drug-resistant cells to conventional therapies. The challenges of toxicity and non-specific targeting associated with some PAK inhibitors, as well as how future approaches for PAK inhibition to overcome these limitations, will also be addressed.
Collapse
|
6
|
Challenging Cutaneous T-Cell Lymphoma: What Animal Models Tell us So Far. J Invest Dermatol 2022; 142:1533-1540. [PMID: 35000751 DOI: 10.1016/j.jid.2021.12.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 12/02/2021] [Accepted: 12/06/2021] [Indexed: 11/22/2022]
Abstract
Cutaneous T-cell lymphomas are characterized by heterogeneity of clinical variants, further complicated by genomic and microenvironmental variables. Furthermore, in vitro experiments are hampered by the low culture efficiency of these malignant cells. Animal models are essential for understanding the pathogenetic mechanisms underlying malignancy and for discovering new anticancer treatments. They are divided into two main categories: those in which tumors arise in the host owing to genetic modifications and those that use tumor cell transplantation. In this review, we summarize the attempts to decipher the complexity of the pathogenesis of cutaneous T-cell lymphoma by exploiting genetically modified and xenograft models.
Collapse
|
7
|
Gu X, Wang Y, Zhang C, Liu Y. GFI-1 overexpression promotes cell proliferation and apoptosis resistance in mycosis fungoides by repressing Bax and P21. Oncol Lett 2021; 22:521. [PMID: 34025788 PMCID: PMC8130034 DOI: 10.3892/ol.2021.12782] [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: 11/21/2020] [Accepted: 03/10/2021] [Indexed: 11/17/2022] Open
Abstract
Mycosis fungoides (MF) is the most common type of cutaneous T-cell lymphoma. The majority of patients with advanced stage MF are resistant to conventional chemotherapy and thus have a poor prognosis. The transcriptional repressor growth factor independence-1 (GFI-1) serves an important role in the development of T-cells. The results of the present study demonstrated that the expression of GFI-1 at different clinical stages of MF was significantly higher compared with benign inflammatory dermatoses, and there was a significant association with disease progression. Gene knockdown of GFI-1 results in the inhibition of Hut-78 cell proliferation and clone formation in vitro, cell cycle arrest and spontaneous apoptosis, upregulation of cell cycle-related P21, as well as the apoptosis-related proteins Bax and Caspase-3, and downregulation of CDK2. Using luciferase assays, and mutational analysis, it was demonstrated that GFI-1 directly regulated the transcription of P21. The results of the present study highlighted a potential molecular therapeutic approach for the treatment of advanced MF.
Collapse
Affiliation(s)
- Xiaoguang Gu
- Department of Dermatology and Venerology, Aviation General Hospital, Beijing 100012, P.R. China.,Institute of Translational Medicine, Chinese Academy of Sciences, Beijing 100012, P.R. China
| | - Yimeng Wang
- Department of Dermatology and Venerology, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Chunlei Zhang
- Department of Dermatology and Venerology, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Yongsheng Liu
- Department of Dermatology and Venerology, Aviation General Hospital, Beijing 100012, P.R. China.,Institute of Translational Medicine, Chinese Academy of Sciences, Beijing 100012, P.R. China
| |
Collapse
|
8
|
Habault J, Thonnart N, Pasquereau-Kotula E, Bagot M, Bensussan A, Villoutreix BO, Marie-Cardine A, Poyet JL. PAK1-Dependent Antitumor Effect of AAC-11‒Derived Peptides on Sézary Syndrome Malignant CD4 + T Lymphocytes. J Invest Dermatol 2021; 141:2261-2271.e5. [PMID: 33745910 DOI: 10.1016/j.jid.2021.01.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/10/2020] [Accepted: 01/08/2021] [Indexed: 01/08/2023]
Abstract
Sézary syndrome is an aggressive form of cutaneous T-cell lymphoma characterized by the presence of a malignant CD4+ T-cell clone in both blood and skin. Its pathophysiology is still poorly understood, and the development of targeted therapies is hampered by the absence of specific target proteins. AAC-11 plays important roles in cancer cell progression and survival and thus has been considered as an anticancer therapeutic target. In this study, we show that a peptide called RT39, comprising a portion of AAC-11‒binding site to its protein partners coupled to the penetratin sequence, induces the specific elimination of the malignant T-cell clone both ex vivo on the circulating cells of patients with Sézary syndrome and in vivo in a subcutaneous xenograft mouse model. RT39 acts by direct binding to PAK1 that is overexpressed, located in the plasma membrane, and constitutively activated in Sézary cells, resulting in their selective depletion by membranolysis. Along with the absence of toxicity, our preclinical efficacy evidence suggests that RT39 might represent a promising alternative therapeutic tool for Sézary syndrome because it spares the nonmalignant immune cells and, contrary to antibody-based immunotherapies, does not require the mobilization of the cellular immunity that shows heavy deficiencies at advanced stages of the disease.
Collapse
Affiliation(s)
- Justine Habault
- Onco-Dermatology and Therapies, INSERM UMRS976, Hôpital Saint Louis, Paris, France; Institut de Recherche Saint Louis, Université de Paris, Paris, France
| | - Nicolas Thonnart
- Onco-Dermatology and Therapies, INSERM UMRS976, Hôpital Saint Louis, Paris, France; Institut de Recherche Saint Louis, Université de Paris, Paris, France
| | - Ewa Pasquereau-Kotula
- Onco-Dermatology and Therapies, INSERM UMRS976, Hôpital Saint Louis, Paris, France; Institut de Recherche Saint Louis, Université de Paris, Paris, France
| | - Martine Bagot
- Onco-Dermatology and Therapies, INSERM UMRS976, Hôpital Saint Louis, Paris, France; Institut de Recherche Saint Louis, Université de Paris, Paris, France; Département de Dermatologie, Hôpital Saint Louis, AP-HP, Paris, France
| | - Armand Bensussan
- Onco-Dermatology and Therapies, INSERM UMRS976, Hôpital Saint Louis, Paris, France; Institut de Recherche Saint Louis, Université de Paris, Paris, France
| | - Bruno O Villoutreix
- INSERM U1141, NeroDiderot, Hôpital Robert-Debré, Paris, France; c-Dithem, Inserm Consortium for Discovery and Innovation in Therapy and Medicine, Paris, France
| | - Anne Marie-Cardine
- Onco-Dermatology and Therapies, INSERM UMRS976, Hôpital Saint Louis, Paris, France; Institut de Recherche Saint Louis, Université de Paris, Paris, France
| | - Jean-Luc Poyet
- Onco-Dermatology and Therapies, INSERM UMRS976, Hôpital Saint Louis, Paris, France; Institut de Recherche Saint Louis, Université de Paris, Paris, France; c-Dithem, Inserm Consortium for Discovery and Innovation in Therapy and Medicine, Paris, France.
| |
Collapse
|
9
|
Up-regulation of P21-activated kinase 1 in osteoarthritis chondrocytes is responsible for osteoarthritic cartilage destruction. Biosci Rep 2021; 40:221716. [PMID: 31868209 PMCID: PMC6954364 DOI: 10.1042/bsr20191017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 11/19/2019] [Accepted: 12/19/2019] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis is mainly caused by a degenerative joint disorder, which is characterized by the gradual degradation of articular cartilage and synovial inflammation. The chondrocyte, the unique resident cell type of articular cartilage, is crucial for the development of osteoarthritis. Previous studies revealed that P21-activated kinase-1 (PAK1) was responsible for the initiation of inflammation. The purpose of the present study was to determine the potential role of PAK1 in osteoarthritis. The level of PAK1 expression was measured by Western blot and quantitative real-time PCR in articular cartilage from osteoarthritis model rats and patients with osteoarthritis. In addition, the functional role of aberrant PAK1 expression was detected in the chondrocytes. We found that the expression of PAK1 was significantly increased in chondrocytes treated with osteoarthritis-related factors. Increased expression of PAK1 was also observed in knee articular cartilage samples from patients with osteoarthritis and osteoarthritis model rats. PAK1 was found to inhibit chondrocytes proliferation and to promote the production of inflammatory cytokines in cartilages chondrocytes. Furthermore, we found that PAK1 modulated the production of extracellular matrix and cartilage degrading enzymes in chondrocytes. Results of the present studies demonstrated that PAK1 might play an important role in the pathogenesis of osteoarthritis.
Collapse
|
10
|
Durgin JS, Weiner DM, Wysocka M, Rook AH. The immunopathogenesis and immunotherapy of cutaneous T cell lymphoma: Pathways and targets for immune restoration and tumor eradication. J Am Acad Dermatol 2021; 84:587-595. [PMID: 33352267 PMCID: PMC7897252 DOI: 10.1016/j.jaad.2020.12.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 11/27/2022]
Abstract
Cutaneous T cell lymphomas (CTCLs) are malignancies of skin-trafficking T cells. Patients with advanced CTCL manifest immune dysfunction that predisposes to infection and suppresses the antitumor immune response. Therapies that stimulate immunity have produced superior progression-free survival compared with conventional chemotherapy, reinforcing the importance of addressing the immune deficient state in the care of patients with CTCL. Recent research has better defined the pathogenesis of these immune deficits, explaining the mechanisms of disease progression and revealing potential therapeutic targets. The features of the malignant cell in mycosis fungoides and Sézary syndrome are now significantly better understood, including the T helper 2 cell phenotype, regulatory T cell cytokine production, immune checkpoint molecule expression, chemokine receptors, and interactions with the microenvironment. The updated model of CTCL immunopathogenesis provides understanding into clinical progression and therapeutic response.
Collapse
Affiliation(s)
- Joseph S Durgin
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David M Weiner
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Maria Wysocka
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alain H Rook
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
11
|
Engin A. Protein Kinase-Mediated Decision Between the Life and Death. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1275:1-33. [PMID: 33539010 DOI: 10.1007/978-3-030-49844-3_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Protein kinases are intracellular signaling enzymes that catalyze the phosphorylation of specific residues in their target substrate proteins. They play important role for regulation of life and death decisions. The complexity of the relationship between death receptors and protein kinases' cell death decision-making mechanisms create many difficulties in the treatment of various diseases. The most of fifteen different cell death pathways, which are reported by Nomenclature Committee on Cell Death (NCCD) are protein kinase signal transduction-mediated negative or positive selections. Tumor necrosis factor (TNF) as a main player of death pathways is a dual-functioning molecule in that it can promote both cell survival or cell death. All apoptotic and necrotic signal transductions are conveyed through death domain-containing death receptors, which are expressed on the surface of nearly all human cells. In humans, eight members of the death receptor family have been identified. While the interaction of TNF with TNF Receptor 1 (TNFR1) activates various signal transduction pathways, different death receptors activate three main signal transduction pathways: nuclear factor kappa B (NF-ĸB)-mediated differentiation or pro-inflammatory cytokine synthesis, mitogen-activated protein kinase (MAPK)-mediated stress response and caspase-mediated apoptosis. The link between the NF-ĸB and the c-Jun NH2-terminal kinase (JNK) pathways comprise another check-point to regulate cell death. TNF-α also promotes the "receptor-interacting serine/threonine protein kinase 1" (RIPK1)/RIPK3/ mixed lineage kinase domain-like pseudokinase (MLKL)-dependent necrosis. Thus, necrosome is mainly comprised of MLKL, RIPK3 and, in some cases, RIPK1. In fact, RIPK1 is at the crossroad between life and death, downstream of various receptors as a regulator of endoplasmic reticulum stress-induced death. TNFR1 signaling complex (TNF-RSC), which contains multiple kinase activities, promotes phosphorylation of transforming growth factor β-activated kinase 1 (TAK1), inhibitor of nuclear transcription factor κB (IκB) kinase (IKK) α/IKKβ, IκBα, and NF-κB. IKKs affect cell-survival pathways in NF-κB-independent manner. Toll-like receptor (TLR) stimulation triggers various signaling pathways dependent on myeloid differentiation factor-88 (MyD88), Interleukin-1 receptor (IL-1R)-associated kinase (IRAK1), IRAK2 and IRAK4, lead to post-translational activation of nucleotide and oligomerization domain (NLRP3). Thereby, cell fate decisions following TLR signaling is parallel with death receptor signaling. Inhibition of IKKα/IKKβ or its upstream activators sensitize cells to death by inducing RIPK1-dependent apoptosis or necroptosis. During apoptosis, several kinases of the NF-κB pathway, including IKK1 and NF-κB essential modulator (NEMO), are cleaved by cellular caspases. This event can terminate the NF-κB-derived survival signals. In both canonical and non-canonical pathways, IKK is key to NF-κB activation. Whereas, the activation process of IKK, the functions of NEMO ubiquitination, IKK-related non-canonical pathway and the nuclear transportation of NEMO and functions of IKKα are still debated in cell death. In addition, cluster of differentiation 95 (CD95)-mediated non-apoptotic signaling and CD95- death-inducing signaling complex (DISC) interactions are waiting for clarification.
Collapse
Affiliation(s)
- Atilla Engin
- Department of General Surgery, Faculty of Medicine, Gazi University, Besevler, Ankara, Turkey.
| |
Collapse
|
12
|
Wang J, Zhu Y, Chen J, Yang Y, Zhu L, Zhao J, Yang Y, Cai X, Hu C, Rosell R, Sun X, Cao P. Identification of a novel PAK1 inhibitor to treat pancreatic cancer. Acta Pharm Sin B 2020; 10:603-614. [PMID: 32322465 PMCID: PMC7161699 DOI: 10.1016/j.apsb.2019.11.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 10/09/2019] [Accepted: 10/15/2019] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer is one of the most aggressive cancers with poor prognosis and a low 5-year survival rate. The family of P21-activated kinases (PAKs) appears to modulate many signaling pathways that contribute to pancreatic carcinogenesis. In this work, we demonstrated that PAK1 is a critical regulator in pancreatic cancer cell growth. PAK1-targeted inhibition is therefore a new potential therapeutic strategy for pancreatic cancer. Our small molecule screening identified a relatively specific PAK1-targeted inhibitor, CP734. Pharmacological and biochemical studies indicated that CP734 targets residue V342 of PAK1 to inhibit its ATPase activity. Further in vitro and in vivo studies elucidated that CP734 suppresses pancreatic tumor growth through depleting PAK1 kinase activity and its downstream signaling pathways. Little toxicity of CP734 was observed in murine models. Combined with gemcitabine or 5-fluorouracil, CP734 also showed synergistic effects on the anti-proliferation of pancreatic cancer cells. All these favorable results indicated that CP734 is a new potential therapeutic candidate for pancreatic cancer.
Collapse
Key Words
- 5-FU, 5-fluorouracil
- ALP, alkaline phosphatase
- ALT, alanine aminotransferase
- ANOVA, analysis of variance
- AST, aspartate aminotransferase
- BCL-2, B-cell lymphoma-2
- BUN, blood urea nitrogen
- CCK-8, cell counting kit-8
- CDC42, cell division cycle 42
- DMEM, Dulbecco's modified Eagle's medium
- DMSO, dimethylsulfoxide
- ERK, extracellular regulated protein kinase
- GEPIA, gene expression profiling interactive analysis
- GTEx, genotype-tissue expression
- Gem, gemcitabine
- HEK293, human embryonic kidney 293
- HTVS, high-throughput virtual screening
- IMEM, improved minimum essential medium
- IP, immunoprecipitation
- Inhibitor
- MEK, mitogen-activated protein kinase kinase
- MEM, modified Eagle's medium
- NSCLC, non-small cell lung cancer
- OHP, oxaliplatin
- OS, overall survival
- PAK, P21-activated kinase
- PAK1
- PARP, poly(ADP-ribose) polymerase
- PAX, paclitaxel
- PSCs, pancreatic stellate cells
- PUMA, P53 upregulated modulator of apoptosis
- PVDF, polyvinylidene fluoride
- Pancreatic cancer
- RAC1, Rac family small GTPase 1
- RIPA, radio immunoprecipitation assay
- RPMI1640, Roswell Park Memorial Institute 1640 medium
- SDS-PAGE, sodium dodecyl sulfate-polyacrylamide gel electrophoresis
- SP, standard precision
- Structure-based virtual screening
- Synergistic effect
- TCGA, The Cancer Genome Atlas
- TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling
- XP, extra precision
Collapse
Affiliation(s)
- Jiaqi Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Yonghua Zhu
- Fullshare Health College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jiao Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Yuhan Yang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Lingxia Zhu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Jiayu Zhao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Yang Yang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Xueting Cai
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Chunping Hu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Rafael Rosell
- Cancer Biology and Precision Medicine Program, Germans Trias i Pujol University Hospital, Badalona, Badalona 08916, Spain
| | - Xiaoyan Sun
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
- Corresponding authors. Tel.: +86 25 85608666; fax: +86 25 52362230.
| | - Peng Cao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Corresponding authors. Tel.: +86 25 85608666; fax: +86 25 52362230.
| |
Collapse
|
13
|
Role and mechanism of matrine alone and combined with acitretin for HaCaT cells and psoriasis-like murine models. Chin Med J (Engl) 2020; 132:2079-2088. [PMID: 31460901 PMCID: PMC6793800 DOI: 10.1097/cm9.0000000000000412] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Acitretin and matrine have been used in the treatment of psoriasis in China. This study was designed to investigate the role and related mechanisms of matrine alone and in combination with acitretin in the treatment of psoriasis in vitro and in vivo. METHODS HaCaT cells were treated with matrine at different concentrations of 0 (blank control), 0.2, 0.4, 0.8, and 1.6 mg/mL for 24, 48, 72 h, respectively. 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium cell viability assay was used to assess the growth and proliferation of HaCaT cells. Cell cycle and apoptosis were detected by flow cytometry. Expression of protein was detected by Western blotting. Autophagy was observed by transmission electron microscopy. Then HaCaT cells were assigned to normal saline (NS) control group, matrine (0.4 mg/mL) group, acitretin (10 μmol/L) group, and matrine plus acitretin group, and the above methods were repeated. In animal experiments, the cumulative score (erythema, scaling, thickening) as a measure of the severity of inflammation was used to measure the skin performance of mice after treated with matrine 50 mg/kg, acitretin 4.5 mg/kg or combination of the two drugs on the psoriasis-like mouse models, respectively. Pathological findings of the lesions were observed, and the protein expressions in the lesions were detected by immunohistochemistry. RESULTS Cell proliferation inhibition was seen in HaCaT cells with treatment of matrine in a dose- and time-dependent manner (P < 0.01, respectively). Cell cycle G0/G1 phase arrest was observed in a dose-dependent way (P < 0.01). The expression of p21 (P < 0.05), LC3II/I (P < 0.01), and Beclin 1 (P < 0.01) increased and the expression of cyclin D1 (P < 0.05) decreased with increasing doses of matrine. Compared with the blank control, more autophagosomes were seen in HaCaT cells treated with matrine at 0.4 mg/mL by transmission electron microscopy (2.667 ± 1.202 vs. 21.33 ± 1.453, t = 9.899, P < 0.01). Cell proliferation inhibition and degree of the G0/G1 phase arrest was significantly higher in matrine plus acitretin group than those in matrine, acitretin, or the NS control group (P < 0.01, respectively). Compared with matrine or acitretin group, the expression of p21 (P < 0.05, P < 0.05) and LC3II/I (P < 0.01, P < 0.05) in matrine plus acitretin group increased significantly and the expression of cyclin D1 (P < 0.01, P < 0.05) and p62 (P < 0.05, P < 0.05) was reduced significantly. Compared with matrine or acitretin, matrine plus acitretin significantly down-regulated the phosphorylation of phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway (P < 0.05) and its downstream p-p70S6K (P < 0.05). In addition, the cumulative score of mice in the matrine plus acitretin group was significantly better than that in the matrine or acitretin group (1.480 ± 0.230 vs. 2.370 ± 0.241, P < 0.01; 1.480 ± 0.230 vs. 2.888 ± 0.341, P < 0.01). The expression of LC3 protein in the matrine plus acitretin group was also higher than that in the matrine, acitretin, or the NS control group (P < 0.05, respectively). CONCLUSIONS Matrine has therapeutic potentials for psoriasis. Matrine and acitretin show synergistic effect via cell cycle arrest and autophagy induction by PI3K/Akt/mTOR pathway.
Collapse
|
14
|
Qi Y, Zhao Y. CBP-triggered KDM2B acetylation accelerates the carcinogenesis of colon cancer. J Cell Physiol 2019; 235:2901-2910. [PMID: 31531877 DOI: 10.1002/jcp.29196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 09/03/2019] [Indexed: 01/08/2023]
Abstract
Lysine (K)-specific demethylase 2B (KDM2B) has been testified to be an oncogene in diverse cancers, which joins in mediating the carcinogenesis of cancers. Nonetheless, the function of KDM2B in colon cancer remains unexplored. The study attempted to disclose the influences of KDM2B acetylation in the progression of colon cancer. SW48 and SUN-C1 cells were transfected with Flag-KDM2B and administrated by trichostatin A and nicotinamide for 24 hr. Immunoprecipitation with a Flag antibody followed by western blot with acetyl-lysine-specific antibody was executed to detect KDM2B acetylation. The correlation between CREB binding protein (CBP) and KDM2B was then investigated. The K-R and K-Q mutants were constructed and the impacts of KDM2B on demethylation of nucleosomal substrates, p21, and puma transcription and the carcinogenesis of colon cancer were probed. CBP immediately evoked KDM2B acetylation at lysine residue 765 in colon cancer cells. Acetylation of KDM2B obviously destroyed the relevance with nucleosomes, demethylation of nucleosomal substrates, and repressed p21 and puma transcription. More important, KDM2B acetylation restrained SUN-C1 cells proliferation and colony formation, meanwhile, hindered cell migration and invasion. Beyond that, the tumor formation was repressed by KDM2B acetylation. The observations testified that CBP-triggered KDM2B acetylation accelerated the carcinogenesis of colon cancer.
Collapse
Affiliation(s)
- Yuxi Qi
- Department of Anus and Intestine Surgery, Jining No.1 People's Hospital, Jining, China.,Affiliated Jining NO.1 People's Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Yanning Zhao
- Department of Anus and Intestine Surgery, Jining No.1 People's Hospital, Jining, China
| |
Collapse
|
15
|
Chen L, Bi S, Hou J, Zhao Z, Wang C, Xie S. Targeting p21-activated kinase 1 inhibits growth and metastasis via Raf1/MEK1/ERK signaling in esophageal squamous cell carcinoma cells. Cell Commun Signal 2019; 17:31. [PMID: 30971268 PMCID: PMC6458688 DOI: 10.1186/s12964-019-0343-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 03/15/2019] [Indexed: 02/06/2023] Open
Abstract
Background p21-activated kinase 1 (PAK1) plays a fundamental role in promoting the development and progression of several cancers and is a potential therapeutic target. However, the biological function and underlying mechanism of PAK1 in esophageal squamous cell carcinoma (ESCC) remain unclear. Methods The expression of PAK1 was detected in both ESCC cell lines and clinical samples. Cell growth was measured by MTT, focus formation and soft agar assays. Cell migration and invasion were detected by wound healing and transwell assays. Animal models of subcutaneous tumourigenicity and tail vein metastasis were performed to determine the inhibitory effect of pharmacological inhibitor IPA-3 on tumor growth and metastasis of ESCC cells. Results We found that PAK1 was frequently overexpressed in ESCC. Ectopic expression of PAK1 promoted cellular growth, colony formation and anchorage-independent growth. Overexpressing PAK1 also enhanced migration, invasion and the expression of MMP-2 and MMP-9 in ESCC cells. In contrast, silencing PAK1 by lentiviral knockdown or a specific inhibitor IPA-3 resulted in a contrary effect. Subsequent investigations revealed that Raf1/MEK1/ERK signaling pathway was involved in PAK1-mediated effect. Enhanced expression of Raf1 attenuated the inhibitory functions of PAK1 shRNA. Whereas blocking of Raf1 by shRNA or specific inhibition of MEK1 by U0126 antagonized the oncogenetic effect of PAK1 on ESCC cells. More importantly, Pharmacological inhibition of PAK1 by IPA-3 significantly suppressed tumor growth and lung metastasis of ESCC cells in vivo. Conclusions These data support that PAK1 is an ideal target for the development of potential therapeutic drugs for ESCC patients even with metastasis. Electronic supplementary material The online version of this article (10.1186/s12964-019-0343-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Liang Chen
- Institute of Chemical Biology, College of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Shuning Bi
- Institute of Chemical Biology, College of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Jiuzhou Hou
- Institute of Chemical Biology, College of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Zhijun Zhao
- Department of Medicine and Therapeutics, Luohe Medical College, Luohe, 462000, China.
| | - Chaojie Wang
- The Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, 475004, China.
| | - Songqiang Xie
- Institute of Chemical Biology, College of Pharmacy, Henan University, Kaifeng, 475004, China.
| |
Collapse
|