1
|
Kumeta M, Otani M, Toyoda M, Yoshimura SH. Acoustic modulation of mechanosensitive genes and adipocyte differentiation. Commun Biol 2025; 8:595. [PMID: 40240542 PMCID: PMC12003795 DOI: 10.1038/s42003-025-07969-1] [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: 08/20/2024] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
Eukaryotic cells are equipped with multiple mechanosensory systems and perceive a wide range of mechanical stimuli from the environment. However, cell-level responses to audible range of acoustic waves, which transmit feeble yet highly frequent physical perturbations, remain largely unexplored. Here, we established a direct sound emission system with a vibrational transducer, and acoustic waves at frequency 440 Hz, 14 kHz, and white noise were transmitted to the murine C2C12 myoblasts at 100 Pa intensity. After 2 and 24 h sound emission, 42 and 145 differentially expressed genes, respectively, were identified using RNA-sequencing. Both cell- and sound-related factors important for inducing gene responses were further investigated. The activation of prostaglandin-endoperoxide synthase 2/cyclooxygenase-2 (Ptgs2/Cox-2), a high and immediate sound-responding gene, is dependent on focal adhesion kinase activation and mediates sound-triggered gene responses by activating prostaglandin E2 synthesis. Adipocyte cells exhibited prominently high sound responses, and their differentiation was significantly suppressed by continuous or periodic acoustic stimulation. Collectively, these findings redefine acoustic waves as cellular stimulators and provide new avenues for applying acoustic techniques in biosciences.
Collapse
Affiliation(s)
- Masahiro Kumeta
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan.
- Center for Living Systems Information Science (CeLiSIS), Kyoto University, Kyoto, Japan.
| | - Makoto Otani
- Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Masahiro Toyoda
- Faculty of Environmental and Urban Engineering, Kansai University, Osaka, Japan
| | - Shige H Yoshimura
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Center for Living Systems Information Science (CeLiSIS), Kyoto University, Kyoto, Japan
| |
Collapse
|
2
|
Tam SW, Cheung AKL, Qin P, Zhang S, Huang Z, Yung KKL. Extracellular Silica Nanomatrices Promote In Vitro Maturation of Anti-tumor Dendritic Cells via Activation of Focal Adhesion Kinase. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2314358. [PMID: 39268785 PMCID: PMC11733713 DOI: 10.1002/adma.202314358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 08/12/2024] [Indexed: 09/15/2024]
Abstract
The efficacy of dendritic cell (DC)-based cancer vaccines is critically determined by the functionalities of in vitro maturated DCs. The maturation of DCs typically relies on chemicals that are cytotoxic or hinder the ability of DCs to efficiently activate the antigen-specific cytotoxic T-lymphocytes (CTLs) against tumors. Herein, the maturation chemicals are replaced with extracellular silica nanomatrices, fabricated by glancing angle deposition, to promote in vitro maturation of murine bone marrow-derived DCs (mBMDCs). The extracellular nanomatrices composed of silica nanozigzags (NZs) enable the generation of mature mBMDCs with upregulated levels of co-stimulatory molecules, C-C chemokine receptor type-7, X-C motif chemokine recetpor-1, DC-specific ICAM-3 grabbing nonintegrin, and enhanced endocytic capacity. The in vitro maturation is partially governed by focal adhesion kinase (FAK) that is mechanically activated in the curved cell adhesions formed at the DC-NZ interfaces. The NZ-maturated mBMDCs can prime the antigen-specific CTLs into programmed cell death protein-1 (PD-1)lowCD44high memory phenotypes in vitro and suppress the growth of tumors in vivo. Meanwhile, the NZ-mediated beneficial effects are also observed in human monocyte-derived DCs. This work demonstrates that the silica NZs promote the anti-tumor capacity of in vitro maturated DCs via the mechanoactivation of FAK, supporting the potential of silica NZs being a promising biomaterial for cancer immunotherapy.
Collapse
Affiliation(s)
- Sze Wah Tam
- Department of BiologyHong Kong Baptist UniversityKowloonHong Kong SARChina
- Golden Meditech Center for NeuroRegeneration SciencesHong Kong Baptist UniversityKowloonHong Kong SARChina
| | | | - Ping Qin
- Department of BiologyHong Kong Baptist UniversityKowloonHong Kong SARChina
- Golden Meditech Center for NeuroRegeneration SciencesHong Kong Baptist UniversityKowloonHong Kong SARChina
| | - Shiqing Zhang
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentJinan UniversityGuangzhou510632China
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug ResearchCollege of PharmacyJinan UniversityGuangzhou510632China
| | - Zhifeng Huang
- Department of ChemistryThe Chinese University of Hong Kong (CUHK)Shatin, N.T.Hong Kong SARChina
- Shenzhen Research Institute of CUHKNo. 10, 2nd Yuexing Road, NanshanShenzhenGuangdong518057China
| | - Ken Kin Lam Yung
- Department of Science and Environmental Studiesthe Education University of Hong KongN.T.Hong Kong SARChina
| |
Collapse
|
3
|
Gao J, Cheng J, Xie W, Zhang P, Liu X, Wang Z, Zhang B. Prospects of focal adhesion kinase inhibitors as a cancer therapy in preclinical and early phase study. Expert Opin Investig Drugs 2024; 33:639-651. [PMID: 38676368 DOI: 10.1080/13543784.2024.2348068] [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: 12/01/2023] [Accepted: 04/23/2024] [Indexed: 04/28/2024]
Abstract
INTRODUCTION FAK, a nonreceptor cytoplasmic tyrosine kinase, plays a crucial role in tumor metastasis, drug resistance, tumor stem cell maintenance, and regulation of the tumor microenvironment. FAK has emerged as a promising target for tumor therapy based on both preclinical and clinical data. AREAS COVERED This paper aims to summarize the molecular mechanisms underlying FAK's involvement in tumorigenesis and progression. Encouraging results have emerged from ongoing clinical trials of FAK inhibitors. Additionally, we present an overview of clinical trials for FAK inhibitors, examining their potential as promising treatments. The pertinent studies gathered from databases including PubMed, ClinicalTrials.gov. EXPERT OPINION Since the first finding in 1990s, targeting FAK has became the focus of interests in many pharmaceutical companies. Through 30 years' discovery, the industry and academy gradually realized the features of FAK target which may not be a driver gene but a solid defense system for the cancer initiation and development. Currently, the ongoing clinical regimens involving FAK inhibition are all the combination strategies in which FAK inhibitors can further strengthen the cancer cell killing effects of other testing agents. The emerging positive signal in clinical trials foresee targeting FAK as class will be an effective mean to fight against cancers.
Collapse
Affiliation(s)
| | | | - Wanyu Xie
- InxMed (Shanghai) Co. Ltd, Shanghai, China
| | - Ping Zhang
- InxMed (Shanghai) Co. Ltd, Shanghai, China
| | - Xuebin Liu
- InxMed (Shanghai) Co. Ltd, Shanghai, China
| | - Zaiqi Wang
- InxMed (Shanghai) Co. Ltd, Shanghai, China
| | | |
Collapse
|
4
|
Borkúti P, Kristó I, Szabó A, Kovács Z, Vilmos P. FERM domain-containing proteins are active components of the cell nucleus. Life Sci Alliance 2024; 7:e202302489. [PMID: 38296350 PMCID: PMC10830384 DOI: 10.26508/lsa.202302489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 01/20/2024] [Accepted: 01/22/2024] [Indexed: 02/05/2024] Open
Abstract
The FERM domain is a conserved and widespread protein module that appeared in the common ancestor of amoebae, fungi, and animals, and is therefore now found in a wide variety of species. The primary function of the FERM domain is localizing to the plasma membrane through binding lipids and proteins of the membrane; thus, for a long time, FERM domain-containing proteins (FDCPs) were considered exclusively cytoskeletal. Although their role in the cytoplasm has been extensively studied, the recent discovery of the presence and importance of cytoskeletal proteins in the nucleus suggests that FDCPs might also play an important role in nuclear function. In this review, we collected data on their nuclear localization, transport, and possible functions, which are still scattered throughout the literature, with special regard to the role of the FERM domain in these processes. With this, we would like to draw attention to the exciting, new dimension of the role of FDCPs, their nuclear activity, which could be an interesting novel direction for future research.
Collapse
Affiliation(s)
| | | | - Anikó Szabó
- HUN-REN Biological Research Centre, Szeged, Hungary
| | - Zoltán Kovács
- HUN-REN Biological Research Centre, Szeged, Hungary
- Doctoral School of Multidisciplinary Medical Science, University of Szeged, Szeged, Hungary
| | - Péter Vilmos
- HUN-REN Biological Research Centre, Szeged, Hungary
| |
Collapse
|
5
|
ZHANG XINFENG, LI SHUANG, SONG MEIRU, CHEN YUE, CHANG LIANGZHENG, LIU ZHERUI, DAI HONGYUAN, WANG YUTAO, YANG GANGQI, JIANG YUN, LU YINYING. Degradation of FAK-targeting by proteolytic targeting chimera technology to inhibit the metastasis of hepatocellular carcinoma. Oncol Res 2024; 32:679-690. [PMID: 38560575 PMCID: PMC10972732 DOI: 10.32604/or.2024.046231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 12/07/2023] [Indexed: 04/04/2024] Open
Abstract
Liver cancer is a prevalent malignant cancer, ranking third in terms of mortality rate. Metastasis and recurrence primarily contribute to the high mortality rate of liver cancer. Hepatocellular carcinoma (HCC) has low expression of focal adhesion kinase (FAK), which increases the risk of metastasis and recurrence. Nevertheless, the efficacy of FAK phosphorylation inhibitors is currently limited. Thus, investigating the mechanisms by which FAK affects HCC metastasis to develop targeted therapies for FAK may present a novel strategy to inhibit HCC metastasis. This study examined the correlation between FAK expression and the prognosis of HCC. Additionally, we explored the impact of FAK degradation on HCC metastasis through wound healing experiments, transwell invasion experiments, and a xenograft tumor model. The expression of proteins related to epithelial-mesenchymal transition (EMT) was measured to elucidate the underlying mechanisms. The results showed that FAK PROTAC can degrade FAK, inhibit the migration and invasion of HCC cells in vitro, and notably decrease the lung metastasis of HCC in vivo. Increased expression of E-cadherin and decreased expression of vimentin indicated that EMT was inhibited. Consequently, degradation of FAK through FAK PROTAC effectively suppressed liver cancer metastasis, holding significant clinical implications for treating liver cancer and developing innovative anti-neoplastic drugs.
Collapse
Affiliation(s)
- XINFENG ZHANG
- The PLA 307 Clinical College of Anhui Medical University, The Fifth Clinical Medical College of Anhui Medical University, Hefei, 230032, China
- Liver Tumor Diagnosis and Research Center, 5th Medical Center of the PLA General Hospital, Beijing, 100039, China
| | - SHUANG LI
- Liver Tumor Diagnosis and Research Center, 5th Medical Center of the PLA General Hospital, Beijing, 100039, China
| | - MEIRU SONG
- The PLA 307 Clinical College of Anhui Medical University, The Fifth Clinical Medical College of Anhui Medical University, Hefei, 230032, China
- Liver Tumor Diagnosis and Research Center, 5th Medical Center of the PLA General Hospital, Beijing, 100039, China
| | - YUE CHEN
- Department of Infection Diseases, Guizhou Medical University, Guiyang, 550025, China
| | - LIANGZHENG CHANG
- Department of Infection Diseases, Guizhou Medical University, Guiyang, 550025, China
| | - ZHERUI LIU
- 302 Clinical Medical School, Peking University, Beijing, China
| | - HONGYUAN DAI
- Department of Infection Diseases, Guizhou Medical University, Guiyang, 550025, China
| | - YUTAO WANG
- 302 Clinical Medical School, Peking University, Beijing, China
| | - GANGQI YANG
- Department of Infection Diseases, Guizhou Medical University, Guiyang, 550025, China
| | - YUN JIANG
- Cell and Gene Therapy Innovation Center, Beijing Lotuslake Biomedical, Science and Technology Park, Beijing, 102206, China
- State Key Laboratory of Chemical Oncogenomics and the Institute of Biopharmaceutical and Health Engineering (iBHE), Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - YINYING LU
- The PLA 307 Clinical College of Anhui Medical University, The Fifth Clinical Medical College of Anhui Medical University, Hefei, 230032, China
- Liver Tumor Diagnosis and Research Center, 5th Medical Center of the PLA General Hospital, Beijing, 100039, China
| |
Collapse
|
6
|
Rashid M, Olson EC. Delayed cortical development in mice with a neural specific deletion of β1 integrin. Front Neurosci 2023; 17:1158419. [PMID: 37250402 PMCID: PMC10213249 DOI: 10.3389/fnins.2023.1158419] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
The adhesion systems employed by migrating cortical neurons are not well understood. Genetic deletion studies of focal adhesion kinase (FAK) and paxillin in mice suggested that these classical focal adhesion molecules control the morphology and speed of cortical neuron migration, but whether β1 integrins also regulate migration morphology and speed is not known. We hypothesized that a β1 integrin adhesion complex is required for proper neuronal migration and for proper cortical development. To test this, we have specifically deleted β1 integrin from postmitotic migrating and differentiating neurons by crossing conditional β1 integrin floxed mice into the NEX-Cre transgenic line. Similar to our prior findings with conditional paxillin deficiency, we found that both homozygous and heterozygous deletion of β1 integrin causes transient mispositioning of cortical neurons in the developing cortex when analyzed pre- and perinatally. Paxillin and β1 integrin colocalize in the migrating neurons and deletion of paxillin in the migrating neuron causes an overall reduction of the β1 integrin immunofluorescence signal and reduction in the number of activated β1 integrin puncta in the migrating neurons. These findings suggest that these molecules may form a functional complex in migrating neurons. Similarly, there was an overall reduced number of paxillin+ puncta in the β1 integrin deficient neurons, despite the normal distribution of FAK and Cx26, a connexin required for cortical migration. The double knockout of paxillin and β1 integrin produces a cortical malpositioning phenotype similar to the paxillin or β1 integrin single knockouts, as would be expected if paxillin and β1 integrin function on a common pathway. Importantly, an isolation-induced pup vocalization test showed that β1 integrin mutants produced a significantly smaller number of calls compared to their littermate controls when analyzed at postnatal day 4 (P4) and revealed a several days trend in reduced vocalization development compared to controls. The current study establishes a role for β1 integrin in cortical development and suggests that β1 integrin deficiency leads to migration and neurodevelopmental delays.
Collapse
Affiliation(s)
- Mamunur Rashid
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, NY, United States
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, United States
| | - Eric C. Olson
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
7
|
Leonov S, Inyang O, Achkasov K, Bogdan E, Kontareva E, Chen Y, Fu Y, Osipov AN, Pustovalova M, Merkher Y. Proteomic Markers for Mechanobiological Properties of Metastatic Cancer Cells. Int J Mol Sci 2023; 24:ijms24054773. [PMID: 36902201 PMCID: PMC10003476 DOI: 10.3390/ijms24054773] [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: 12/12/2022] [Revised: 01/26/2023] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
The major cause (more than 90%) of all cancer-related deaths is metastasis, thus its prediction can critically affect the survival rate. Metastases are currently predicted by lymph-node status, tumor size, histopathology and genetic testing; however, all these are not infallible, and obtaining results may require weeks. The identification of new potential prognostic factors will be an important source of risk information for the practicing oncologist, potentially leading to enhanced patient care through the proactive optimization of treatment strategies. Recently, the new mechanobiology-related techniques, independent of genetics, based on the mechanical invasiveness of cancer cells (microfluidic, gel indentation assays, migration assays etc.), demonstrated a high success rate for the detection of tumor cell metastasis propensity. However, they are still far away from clinical implementation due to complexity. Hence, the exploration of novel markers related to the mechanobiological properties of tumor cells may have a direct impact on the prognosis of metastasis. Our concise review deepens our knowledge of the factors that regulate cancer cell mechanotype and invasion, and incites further studies to develop therapeutics that target multiple mechanisms of invasion for improved clinical benefit. It may open a new clinical dimension that will improve cancer prognosis and increase the effectiveness of tumor therapies.
Collapse
Affiliation(s)
- Sergey Leonov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Moscow Region, Russia
- Institute of Cell Biophysics, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia
| | - Olumide Inyang
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Moscow Region, Russia
| | - Konstantin Achkasov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Moscow Region, Russia
| | - Elizaveta Bogdan
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Moscow Region, Russia
| | - Elizaveta Kontareva
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Moscow Region, Russia
| | - Yongheng Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ying Fu
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Andreyan N. Osipov
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical-Biological Agency, 123098 Moscow, Russia
- N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119991 Moscow, Russia
- Correspondence:
| | - Margarita Pustovalova
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Moscow Region, Russia
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical-Biological Agency, 123098 Moscow, Russia
| | - Yulia Merkher
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Moscow Region, Russia
| |
Collapse
|
8
|
Simiate and the focal adhesion kinase FAK1 cooperate in the regulation of dendritogenesis. Sci Rep 2022; 12:11274. [PMID: 35787638 PMCID: PMC9253104 DOI: 10.1038/s41598-022-14460-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/07/2022] [Indexed: 11/09/2022] Open
Abstract
Despite the crucial importance of dendritogenesis for the correct functioning of neurons, the molecular mechanisms underlying neuronal arborisation are still not well understood. Current models suggest that distinct parts and phases of dendritic development are regulated by the expression of distinct transcription factors, that are able to target the cytoskeleton. Two proteins recently implicated in dendritogenesis are the Focal Adhesion Kinase FAK1 and the Actin-binding protein Simiate. Using heterologous expression systems as well as mouse brain extracts in combination with coprecipitation assays, we show that Simiate is able to associate with FAK1. Differential centrifugation experiments further revealed the interaction to be present in cytosolic as well as nuclear fractions. Inside the nucleus though, Simiate preferentially binds to a FAK1 isoform of 80 kDa, which has previously been shown to regulate transcription factor activity. Investigating the function of both proteins in primary hippocampal cultures, we further found that FAK1 and Simiate have distinct roles in dendritogenesis: While FAK1 increases dendrite length and number, Simiate preferentially enhances growth and branching. However, if being confined to the nucleus, Simiate selectively triggers primary dendrite formation, enhancing transcription activity at the same time. Since the effect on primary dendrites is specifically re-normalized by a co-expression of FAK1 and Simiate in the nucleus, the data implies that the two proteins interact to counterbalance each other in order to control dendrite formation. Looking at the role of the cytosolic interaction of FAK1 and Simiate, we found that neurotrophin induced dendritogenesis causes a striking colocalisation of FAK1 and Simiate in dendritic growth cones, which is not present otherwise, thus suggesting that the cytosolic interaction stimulates growth cone mediated dendritogenesis in response to certain external signals. Taken together, the data show that FAK1 and Simiate exert several and distinct actions during the different phases of dendritogenesis and that these actions are related to their subcellular localisation and their interaction.
Collapse
|
9
|
D'Amico G, Fernandez I, Gómez-Escudero J, Kim H, Maniati E, Azman MS, Mardakheh FK, Serrels B, Serrels A, Parsons M, Squire A, Birdsey GM, Randi AM, Bolado-Carrancio A, Gangeswaran R, Reynolds LE, Bodrug N, Wang Y, Wang J, Meier P, Hodivala-Dilke KM. ERG activity is regulated by endothelial FAK coupling with TRIM25/USP9x in vascular patterning. Development 2022; 149:dev200528. [PMID: 35723257 PMCID: PMC9340553 DOI: 10.1242/dev.200528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/29/2022] [Indexed: 11/20/2022]
Abstract
Precise vascular patterning is crucial for normal growth and development. The ERG transcription factor drives Delta-like ligand 4 (DLL4)/Notch signalling and is thought to act as a pivotal regulator of endothelial cell (EC) dynamics and developmental angiogenesis. However, molecular regulation of ERG activity remains obscure. Using a series of EC-specific focal adhesion kinase (FAK)-knockout (KO) and point-mutant FAK-knock-in mice, we show that loss of ECFAK, its kinase activity or phosphorylation at FAK-Y397, but not FAK-Y861, reduces ERG and DLL4 expression levels together with concomitant aberrations in vascular patterning. Rapid immunoprecipitation mass spectrometry of endogenous proteins identified that endothelial nuclear-FAK interacts with the deubiquitinase USP9x and the ubiquitin ligase TRIM25. Further in silico analysis confirms that ERG interacts with USP9x and TRIM25. Moreover, ERG levels are reduced in FAKKO ECs via a ubiquitin-mediated post-translational modification programme involving USP9x and TRIM25. Re-expression of ERG in vivo and in vitro rescues the aberrant vessel-sprouting defects observed in the absence of ECFAK. Our findings identify ECFAK as a regulator of retinal vascular patterning by controlling ERG protein degradation via TRIM25/USP9x.
Collapse
Affiliation(s)
- Gabriela D'Amico
- Centre for Tumour Microenvironment, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Isabelle Fernandez
- Centre for Tumour Microenvironment, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Jesús Gómez-Escudero
- Centre for Tumour Microenvironment, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Hyojin Kim
- The Breakthrough Toby Robins Breast Cancer Research Centre, Institute of Cancer Research, Chester Beatty Laboratories, Fulham Road, London SW3 6JB, UK
| | - Eleni Maniati
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Muhammad Syahmi Azman
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Faraz K. Mardakheh
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Bryan Serrels
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden G61 1QH, UK
| | - Alan Serrels
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Maddy Parsons
- Kings College London, Randall Centre of Cell and Molecular Biophysics, Room 3.22B, New Hunts House, Guys Campus, London SE1 1UL, UK
| | - Anthony Squire
- IMCES - Imaging Centre Essen, Institute for Experimental Immunology and Imaging, University Clinic Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Graeme M. Birdsey
- National Heart & Lung Institute, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - Anna M. Randi
- National Heart & Lung Institute, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | | | - Rathi Gangeswaran
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Louise E. Reynolds
- Centre for Tumour Microenvironment, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Natalia Bodrug
- Centre for Tumour Microenvironment, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Yaohe Wang
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Jun Wang
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Pascal Meier
- The Breakthrough Toby Robins Breast Cancer Research Centre, Institute of Cancer Research, Chester Beatty Laboratories, Fulham Road, London SW3 6JB, UK
| | - Kairbaan M. Hodivala-Dilke
- Centre for Tumour Microenvironment, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
10
|
Alba A, Villaggio G, Messina GML, Caruso M, Federico C, Cambria MT, Marletta G, Sinatra F. Cytostatic Effects of Polyethyleneimine Surfaces on the Mesenchymal Stromal Cell Cycle. Polymers (Basel) 2022; 14:polym14132643. [PMID: 35808689 PMCID: PMC9269326 DOI: 10.3390/polym14132643] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 02/04/2023] Open
Abstract
Polyelectrolytes assembled layer-by-layer (PEMs) are commonly used as functional coatings to build-up biological interfaces, particularly suitable as compatible layers for the interaction with a biological medium, providing suitable conditions to promote or prevent cell seeding while maintaining the phenotype. The proper assessment of the biocompatibility of PEMs and the elucidation of the related mechanisms are therefore of paramount importance. In this study, we report in detail the effect of two different PEM endings, polystyrene sulfonate (PSS) and polyethylenimine (PEI), respectively, on the cell adhesion, growth, and viability of human bone mesenchymal stromal cells (MSCs). The results have shown that PSS-ended substrates appear to be the most suitable to drive the cell adhesion and phenotype maintenance of MSCs, showing good biocompatibility. On the contrary, while the cells seem to adhere more quickly and strongly on the PEI-ended surfaces, the interaction with PEI significantly affects the growth and viability, reducing the cell spreading capability, by sequestering the adhesion molecules already in the very early steps of cell–substrate contact. These results point to the promotion of a cytostatic effect of PEI, rather than the often-claimed cytotoxicity.
Collapse
Affiliation(s)
- Anna Alba
- Section of Biology and Genetic, Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia, 65, 95123 Catania, Italy; (A.A.); (G.V.)
| | - Giusy Villaggio
- Section of Biology and Genetic, Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia, 65, 95123 Catania, Italy; (A.A.); (G.V.)
| | - Grazia Maria Lucia Messina
- Laboratory for Molecular Surface and Nanotechnology (LAMSUN), Department of Chemical Sciences, University of Catania and CSGI, Viale A. Doria, 6, 95125 Catania, Italy;
- Correspondence: (G.M.L.M.); (F.S.); Tel.: +39-095-7385083 (G.M.L.M.)
| | - Massimo Caruso
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia, 65, 95123 Catania, Italy; (M.C.); (M.T.C.)
| | - Concetta Federico
- Section of Animal Biology, Department of Biological, Geological and Environmental Sciences, University of Catania, Via Androne, 81, 95124 Catania, Italy;
| | - Maria Teresa Cambria
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia, 65, 95123 Catania, Italy; (M.C.); (M.T.C.)
| | - Giovanni Marletta
- Laboratory for Molecular Surface and Nanotechnology (LAMSUN), Department of Chemical Sciences, University of Catania and CSGI, Viale A. Doria, 6, 95125 Catania, Italy;
| | - Fulvia Sinatra
- Section of Biology and Genetic, Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia, 65, 95123 Catania, Italy; (A.A.); (G.V.)
- Correspondence: (G.M.L.M.); (F.S.); Tel.: +39-095-7385083 (G.M.L.M.)
| |
Collapse
|
11
|
Meng F, Zhang K, Yang C, Zhang K, Xu Q, Ren R, Zhou Y, Sun Y, Peng Y, Li Y, Guo H, Ren Y, Zhao Z. Prognostic Pathways Guide Drug Indications in Pan-Cancers. Front Oncol 2022; 12:849552. [PMID: 35372084 PMCID: PMC8964428 DOI: 10.3389/fonc.2022.849552] [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: 01/06/2022] [Accepted: 02/07/2022] [Indexed: 11/18/2022] Open
Abstract
Pathway-level analysis is a powerful approach enabling the interpretation of post-genomic data at a higher level than that of individual molecules. Molecular-targeted therapy focusing on cascade signaling pathways has become a new paradigm in anticancer therapy, instead of a single protein. However, the approaches to narrowing down the long list of biological pathways are limited. Here, we proposed a strategy for in silico Drug Prescription on biological pathways across pan-Cancers (CDP), by connecting drugs to candidate pathways. Applying on a list of 120 traditional Chinese medicines (TCM), we especially identified the “TCM–pathways–cancers” triplet and constructed it into a heterogeneous network across pan-cancers. Applying them into TCMs, the computational prescribing methods deepened the understanding of the efficacy of TCM at the molecular level. Further applying them into Western medicines, CDP could promote drug reposition avoiding time-consuming developments of new drugs.
Collapse
Affiliation(s)
- Fanlin Meng
- Marketing and Management Department, CapitalBio Technology, Beijing, China.,National Engineering Research Center for Beijing Biochip Technology, Beijing, China
| | - Kenan Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Changlin Yang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Ke Zhang
- National Engineering Research Center for Beijing Biochip Technology, Beijing, China
| | - Quan Xu
- National Engineering Research Center for Beijing Biochip Technology, Beijing, China
| | - Ruifang Ren
- National Engineering Research Center for Beijing Biochip Technology, Beijing, China
| | - Yiming Zhou
- National Engineering Research Center for Beijing Biochip Technology, Beijing, China
| | - Yimin Sun
- Marketing and Management Department, CapitalBio Technology, Beijing, China.,National Engineering Research Center for Beijing Biochip Technology, Beijing, China
| | - Yan Peng
- Marketing and Management Department, CapitalBio Technology, Beijing, China
| | - Yanze Li
- Marketing and Management Department, CapitalBio Technology, Beijing, China
| | - Hongyan Guo
- National Engineering Research Center for Beijing Biochip Technology, Beijing, China
| | - Yonghong Ren
- Marketing and Management Department, CapitalBio Technology, Beijing, China
| | - Zheng Zhao
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| |
Collapse
|
12
|
Gu W, Zhang L, Zhang X, Wang B, Shi X, Hu K, Ye Y, Liu G. MiR-15p-5p Mediates the Coordination of ICAM-1 and FAK to Promote Endothelial Cell Proliferation and Migration. Inflammation 2022; 45:1402-1417. [PMID: 35079920 DOI: 10.1007/s10753-022-01630-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 11/25/2022]
Abstract
Intercellular adhesion molecule-1 (ICAM-1) in endothelial cells is critical for neutrophil adhesion and transmigration across the endothelium. Focal adhesion kinase (FAK), which controls the turnover of focal adhesion to regulate cell adhesion and migration, plays a role in the resolution of inflammation. However, the coordinated involvement of ICAM-1 and FAK during endothelial inflammation has yet to be elucidated. This study reports that, as part of an inflammatory response, ICAM-1 controls FAK expression in endothelial cells via the microRNA miR-15b-5p. Induction of lung injury by lipopolysaccharide (LPS) resulted in higher levels of FAK expression in inflammatory tissues, while in ICAM-1 knockout mice, FAK expression was reduced in the lungs. FAK expression was also reduced in endothelial cells following ICAM-1 siRNA downregulation. Furthermore, ICAM-1 inhibited miR-15b-5p expression while increasing FAK mRNA and protein expression via binding of miR-15b-5p to the 3' untranslated region (UTR) of FAK. ICAM-1 inhibited miR-15b-5p promoter activity and hence reduced miR-15b-5p expression. FAK increased endothelial cell proliferation and migration, whereas miR-15b-5p inhibited cell proliferation and migration. These findings indicate that the inflammatory molecule ICAM-1 regulates FAK expression via miR-15b-5p levels, which in turn controls endothelial cell proliferation and migration.
Collapse
Affiliation(s)
- Wei Gu
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, 2600 Donghai StreetAnhui Province, Bengbu, 233030, China
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui Province, Bengbu, 233030, China
| | - Li Zhang
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, 2600 Donghai StreetAnhui Province, Bengbu, 233030, China
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui Province, Bengbu, 233030, China
| | - Xinhua Zhang
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Hubei Province, Wuhan, 430070, China
| | - Binyu Wang
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Hubei Province, Wuhan, 430070, China
| | - Xiaoyu Shi
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui Province, Bengbu, 233030, China
| | - Kang Hu
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, 2600 Donghai StreetAnhui Province, Bengbu, 233030, China
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui Province, Bengbu, 233030, China
| | - Yingying Ye
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, 2600 Donghai StreetAnhui Province, Bengbu, 233030, China
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui Province, Bengbu, 233030, China
| | - Guoquan Liu
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, 2600 Donghai StreetAnhui Province, Bengbu, 233030, China.
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui Province, Bengbu, 233030, China.
| |
Collapse
|
13
|
Design, synthesis and biological evaluation of 7-((7H-pyrrolo[2,3-d]pyrimidin-4-yl)oxy)-2,3-dihydro-1H-inden-1-one derivatives as potent FAK inhibitors for the treatment of ovarian cancer. Eur J Med Chem 2022; 228:113978. [PMID: 34810020 DOI: 10.1016/j.ejmech.2021.113978] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 02/05/2023]
Abstract
Focal adhesion kinase (FAK) promotes tumor progression by intracellular signal transduction and regulation of gene expression and protein turnover, which is a compelling therapeutic target for various cancer types, including ovarian cancer. However, the clinical responses of FAK inhibitors remain unsatisfactory. Here, we describe the discovery of FAK inhibitors using a scaffold hopping strategy. Structure-activity relationship (SAR) exploration identified 36 as a potent FAK inhibitor, which exhibited inhibitory activities against FAK signaling in vitro. Treatment with 36 not only decreased migration and invasion of PA-1 cells, but also reduced expression of MMP-2 and MMP-9. Moreover, 36 inhibited tumor growth and metastasis, and no obvious adverse effects were observed during the in vivo study. These results revealed the potential of FAK inhibitor 36 for treatment of ovarian cancer.
Collapse
|
14
|
Quispe PA, Lavecchia MJ, León IE. Focal adhesion kinase inhibitors in the treatment of solid tumors: Preclinical and clinical evidence. Drug Discov Today 2021; 27:664-674. [PMID: 34856395 DOI: 10.1016/j.drudis.2021.11.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/21/2021] [Accepted: 11/23/2021] [Indexed: 01/25/2023]
Abstract
Focal Adhesion Kinase (FAK) is a 125-kDa cytoplasmic protein kinase that is implicated in several cellular functions. This protein is an attractive molecular target for cancer therapy because a wide variety of studies have demonstrated associations between the activation or elevated expression of FAK and tumor progression, invasion, and drug resistance in malignant tumors. Here, we review the strategies used to inhibit FAK activity in solid tumors. We also include an overview of the preclinical (in vitro and in vivo) and clinical studies on FAK inhibitors.
Collapse
Affiliation(s)
- Patricia A Quispe
- Centro de Química Inorgánica (CEQUINOR, CONICET-UNLP), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Bv 120 1465, 1900 La Plata, Argentina
| | - Martin J Lavecchia
- Centro de Química Inorgánica (CEQUINOR, CONICET-UNLP), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Bv 120 1465, 1900 La Plata, Argentina.
| | - Ignacio E León
- Centro de Química Inorgánica (CEQUINOR, CONICET-UNLP), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Bv 120 1465, 1900 La Plata, Argentina.
| |
Collapse
|
15
|
Focal adhesion kinase splicing and protein activation in papillary thyroid carcinoma progression. Histochem Cell Biol 2021; 157:183-194. [PMID: 34817652 DOI: 10.1007/s00418-021-02056-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2021] [Indexed: 01/03/2023]
Abstract
Papillary thyroid carcinoma (PTC), a common endocrine malignancy, presents a challenge from a prognostic standpoint. Molecular alterations underlying PTC progression include deregulation of focal adhesion kinase (FAK) at post-transcriptional and post-translational levels. Searching for candidate markers of PTC progression, we investigated the prognostic significance of FAK alterations on mRNA/protein level. The expression levels and subcellular localisation of auto-phosphorylated FAK (pY397-FAK) were determined by western blot (WB) and immunohistochemistry. The quantity of total FAK mRNA, alternatively spliced FAK-Del26 and FAK-Del33 variants were analysed by RT-qPCR and related to pY397-FAK expression and subcellular distribution. The results were correlated with clinicopathological parameters of the patients. The expression of pY397-FAK was significantly elevated in malignant samples. Active FAK showed predominant cytoplasmic distribution with co-occurrence along the membrane, while nuclear staining was found less frequently. Expression of pY397-FAK in separate cellular compartments correlated with adverse clinicopathological parameters, but the strongest association was found when their mean scores were calculated. Alternatively spliced FAK-Del33 and total FAK transcripts positively correlated to pY397-FAK protein levels as well as to characteristics of PTC advancement. Over-expression of FAK on mRNA (total and Del-33) and activated protein (pY397-FAK) levels is a feature of PTC advanced stages. Of the analysed alterations, the mean pY397-FAK IHC score showed the best predictive performance. Correlation between mRNA FAK-Del33 and pY397-FAK expression implies a regulatory role of alternative splicing in PTC patients.
Collapse
|
16
|
Liu ZZ(G, Taiyab A, West-Mays JA. MMP9 Differentially Regulates Proteins Involved in Actin Polymerization and Cell Migration during TGF-β-Induced EMT in the Lens. Int J Mol Sci 2021; 22:ijms222111988. [PMID: 34769418 PMCID: PMC8584335 DOI: 10.3390/ijms222111988] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/23/2021] [Accepted: 11/02/2021] [Indexed: 11/16/2022] Open
Abstract
Fibrotic cataracts have been attributed to transforming growth factor-beta (TGF-β)-induced epithelial-to-mesenchymal transition (EMT). Using mouse knockout (KO) models, our laboratory has identified MMP9 as a crucial protein in the TGF-β-induced EMT process. In this study, we further revealed an absence of alpha-smooth muscle actin (αSMA) and filamentous-actin (F-actin) stress fibers in MMP9KO mouse lens epithelial cell explants (LECs). Expression analysis using NanoString revealed no marked differences in αSMA (ACTA2) and beta-actin (β-actin) (ACTB) mRNA between the lenses of TGF-β-overexpressing (TGF-βtg) mice and TGF-βtg mice on a MMP9KO background. We subsequently conducted a protein array that revealed differential regulation of proteins known to be involved in actin polymerization and cell migration in TGF-β-treated MMP9KO mouse LECs when compared to untreated controls. Immunofluorescence analyses using rat LECs and the novel MMP9-specific inhibitor, JNJ0966, revealed similar differential regulation of cortactin, FAK, LIMK1 and MLC2 as observed in the array. Finally, a reduction in the nuclear localization of MRTF-A, a master regulator of cytoskeletal remodeling during EMT, was observed in rat LECs co-treated with JNJ0966 and TGF-β. In conclusion, MMP9 deficiency results in differential regulation of proteins involved in actin polymerization and cell migration, and this in turn prevents TGF-β-induced EMT in the lens.
Collapse
Affiliation(s)
| | | | - Judith A. West-Mays
- Correspondence: ; Tel.: +1-(905)-525-9140 (ext. 26237); Fax: +1-(905)-525-7400
| |
Collapse
|
17
|
Anderson EM, Thomassian S, Gong J, Hendifar A, Osipov A. Advances in Pancreatic Ductal Adenocarcinoma Treatment. Cancers (Basel) 2021; 13:5510. [PMID: 34771675 PMCID: PMC8583016 DOI: 10.3390/cancers13215510] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/26/2021] [Accepted: 10/26/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is one of the deadliest malignancies among all cancers. Despite curative intent, surgery and the use of standard cytotoxic chemotherapy and radiation therapy, PDAC remains treatment-resistant. In recent years, more contemporary treatment modalities such as immunotherapy via checkpoint inhibition have shown some promise in many other malignancies, yet PDAC still eludes an effective curative treatment. In investigating these phenomena, research has suggested that the significant desmoplastic and adaptive tumor microenvironment (TME) of PDAC promote the proliferation of immunosuppressive cells and act as major obstacles to treatment efficacy. In this review, we explore challenges associated with the treatment of PDAC, including its unique immunosuppressive TME. This review examines the role of surgery in PDAC, recent advances in surgical approaches and surgical optimization. We further focus on advances in immunotherapeutic approaches, including checkpoint inhibition, CD40 agonists, and discuss promising immune-based future strategies, such as therapeutic neoantigen cancer vaccines as means of overcoming the resistance mechanisms which underly the dense stroma and immune milieu of PDAC. We also explore unique signaling, TME and stromal targeting via novel small molecule inhibitors, which target KRAS, FAK, CCR2/CCR5, CXCR4, PARP and cancer-associated fibroblasts. This review also explores the most promising strategy for advancement in treatment of pancreatic cancer by reviewing contemporary combinatorial approaches in efforts to overcome the treatment refractory nature of PDAC.
Collapse
Affiliation(s)
- Eric M. Anderson
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Shant Thomassian
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; (S.T.); (J.G.); (A.H.)
| | - Jun Gong
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; (S.T.); (J.G.); (A.H.)
| | - Andrew Hendifar
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; (S.T.); (J.G.); (A.H.)
| | - Arsen Osipov
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; (S.T.); (J.G.); (A.H.)
| |
Collapse
|
18
|
Hernández-Bule ML, Toledano-Macías E, Naranjo A, de Andrés-Zamora M, Úbeda A. In vitro stimulation with radiofrequency currents promotes proliferation and migration in human keratinocytes and fibroblasts. Electromagn Biol Med 2021; 40:338-352. [PMID: 34315307 DOI: 10.1080/15368378.2021.1938113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Capacitive-resistive electric transfer (CRET) therapies have been proposed as strategies for regeneration of cutaneous tissue lesions. Previous studies by our group have shown that intermittent stimulation with 448 kHz CRET currents at subthermal densities promotes in vitro proliferation of human stem cells involved in tissue regeneration. The present study investigates the effects of the in vitro exposure to these radiofrequency (RF) currents on the proliferation and migration of keratinocytes and fibroblasts, the main cell types involved in skin regeneration. The effects of the electric stimulation on cell proliferation and migration were studied through XTT and wound closure assays, respectively. The CRET effects on the expression and location of proteins involved in proliferation and migration were assessed by immunoblot and immunofluorescence. The obtained results reveal that electrostimulation promotes proliferation and/or migration in keratinocytes and fibroblasts. These effects would be mediated by changes observed in the expression and location of intercellular adhesion proteins such as β-catenin and E-cadherin, of proteins involved in cell-to-substrate adhesion such as vinculin, p-FAK and the metalloproteinase MMP-9, and of other proteins that control both processes: MAP kinases p-p38, p-JUNK and p-ERK1/2. These responses could represent a mechanism underlying the promotion of normotrophic wound regeneration induced by CRET. Indeed, electric stimulation would favor completion of granulation tissue formation prior to the closure of the outer tissue layers, thus preventing abnormal wound cicatrization or chronification.
Collapse
Affiliation(s)
| | - Elena Toledano-Macías
- Servicio de Bioelectromagnetismo, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Aida Naranjo
- Departamento de Ingeniería Eléctrica, Escuela Técnica Superior de Ingeniería y Diseño Industrial, Universidad Politécnica de Madrid, Madrid, Spain
| | - Marina de Andrés-Zamora
- Departamento de Ingeniería Eléctrica, Escuela Técnica Superior de Ingeniería y Diseño Industrial, Universidad Politécnica de Madrid, Madrid, Spain
| | - Alejandro Úbeda
- Servicio de Bioelectromagnetismo, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| |
Collapse
|
19
|
Gilardi M, Bersini S, Valtorta S, Proietto M, Crippa M, Boussommier-Calleja A, Labelle M, Moresco RM, Vanoni M, Kamm RD, Moretti M. The driving role of the Cdk5/Tln1/FAK S732 axis in cancer cell extravasation dissected by human vascularized microfluidic models. Biomaterials 2021; 276:120975. [PMID: 34333365 DOI: 10.1016/j.biomaterials.2021.120975] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 06/04/2021] [Accepted: 06/11/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Understanding the molecular mechanisms of metastatic dissemination, the leading cause of death in cancer patients, is required to develop novel, effective therapies. Extravasation, an essential rate-limiting process in the metastatic cascade, includes three tightly coordinated steps: cancer cell adhesion to the endothelium, trans-endothelial migration, and early invasion into the secondary site. Focal adhesion proteins, including Tln1 and FAK, regulate the cytoskeleton dynamics: dysregulation of these proteins is often associated with metastatic progression and poor prognosis. METHODS Here, we studied the previously unexplored role of these targets in each extravasation step using engineered 3D in vitro models, which recapitulate the physiological vascular niche experienced by cancer cells during hematogenous metastasis. RESULTS Human breast cancer and fibrosarcoma cell lines respond to Cdk5/Tln1/FAK axis perturbation, impairing their metastatic potential. Vascular breaching requires actin polymerization-dependent invadopodia formation. Invadopodia generation requires the structural function of FAK and Tln1 rather than their activation through phosphorylation. Our data support that the inhibition of FAKS732 phosphorylation delocalizes ERK from the nucleus, decreasing ERK phosphorylated form. These findings indicate the critical role of these proteins in driving trans-endothelial migration. In fact, both knock-down experiments and chemical inhibition of FAK dramatically reduces lung colonization in vivo and TEM in microfluidic setting. Altogether, these data indicate that engineered 3D in vitro models coupled to in vivo models, genetic, biochemical, and imaging tools represent a powerful weapon to increase our understanding of metastatic progression. CONCLUSIONS These findings point to the need for further analyses of previously overlooked phosphorylation sites of FAK, such as the serine 732, and foster the development of new effective antimetastatic treatments targeting late events of the metastatic cascade.
Collapse
Affiliation(s)
- Mara Gilardi
- Cell and Tissue Engineering Lab, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy; Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126, Milan, Italy; Institute of Pathology, University Hospital Basel, University of Basel, 4031, Basel, Switzerland.
| | - Simone Bersini
- Cell and Tissue Engineering Lab, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy; Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale, Lugano, Switzerland.
| | - Silvia Valtorta
- Università Degli Studi di Milano-Bicocca, Department of Medicine and Surgery and Tecnomed Foundation, Monza, Italy; Institute of Bioimaging and Molecular Physiology of National Researches Council (IBFM-CNR), Segrate, Italy.
| | - Marco Proietto
- Department of Biology-University of California - San Diego, La Jolla, CA, USA.
| | - Martina Crippa
- Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale, Lugano, Switzerland; Laboratory of Biological Structures Mechanics, Chemistry, Material and Chemical Engineering Department "Giulio Natta", Politecnico di Milano, Milan, Italy.
| | - Alexandra Boussommier-Calleja
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA.
| | - Myriam Labelle
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Rosa Maria Moresco
- Università Degli Studi di Milano-Bicocca, Department of Medicine and Surgery and Tecnomed Foundation, Monza, Italy; Institute of Bioimaging and Molecular Physiology of National Researches Council (IBFM-CNR), Segrate, Italy.
| | - Marco Vanoni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126, Milan, Italy; ISBE.IT/ Centre of Systems Biology, Milano, Italy.
| | - Roger D Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA.
| | - Matteo Moretti
- Cell and Tissue Engineering Lab, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy; Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale, Lugano, Switzerland; Euler Institute, Biomedical Sciences Faculty, Università Della Svizzera Italiana, Lugano, Switzerland.
| |
Collapse
|
20
|
Brullo C, Tasso B. New Insights on Fak and Fak Inhibitors. Curr Med Chem 2021; 28:3318-3338. [PMID: 33143618 DOI: 10.2174/0929867327666201103162239] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 09/08/2020] [Accepted: 09/19/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Focal adhesion kinase (Fak) is a cytoplasmic protein tyrosine kinase overexpressed and activated in different solid cancers; it has shown an important role in metastasis formation, cell migration, invasion and angiogenesis and consequently it has been proposed as a potential target in cancer therapy, particularly in a metastatic phase. In recent years, different investigations have highlighted the importance of new Fak inhibitors as potential anti-cancer drugs, but other studies evidenced its role in different pathologies related to the cardiac function or viral infection. METHODS An extensive bibliographic research (104 references) has been done concerning the structure of Fak, its importance in tumor development, but also in other pathologies currently under study. The compounds currently subjected to clinical studies were therefore treated using the appropriate databases. Finally, the main chemical scaffolds currently under preclinical investigation were analyzed, focusing on their molecular structures and on the activity structure relationships (SAR). RESULTS At the moment, only a few reversible ATP-competitive inhibitors are under investigation in pre-clinical studies and clinical trials. Other compounds, with different chemical scaffolds, are investigated to obtain more active and selective Fak inhibitors. This mini-review is a summary of different Fak functions in cancer and other pathologies; the compounds today in clinical trials and the recent chemical scaffolds (also included in patents) giving the most interesting results are investigated. In addition, PROTAC molecules are reported. CONCLUSION All reported results evidenced that additional studies are necessary to design and synthesize new selective and more active compounds, although promising information has been obtained from associations between Fak inhibitors and other different anti- cancer drugs. In addition, the other important roles evidenced, both at the nuclear level and in non-cancerous cells, make this protein an increasingly important target in pharmaceutical chemistry.
Collapse
Affiliation(s)
- Chiara Brullo
- Department of Pharmacy, University of Genova, Viale Benedetto XV, 3-I16132 Genova, Italy
| | - Bruno Tasso
- Department of Pharmacy, University of Genova, Viale Benedetto XV, 3-I16132 Genova, Italy
| |
Collapse
|
21
|
SERINE-910 Phosphorylated Focal Adhesion Kinase Expression Predicts Better Overall and Disease-free Survival in Melanoma. Appl Immunohistochem Mol Morphol 2020; 28:130-138. [PMID: 32044881 DOI: 10.1097/pai.0000000000000744] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Focal adhesion kinase (FAK) is a cytoplasmic tyrosine kinase that mediates multiple cellular functions such as survival, invasion, and migration. FAK has been found to be over-expressed in various human cancers, including melanoma. FAK molecule has several tyrosine, serine, and threonine phosphorylation sites which have an important regulatory role. Tyrosine phosphorylation of FAK has been extensively studied, however little is known about the role of serine phosphorylation. We sought to examine the frequency and extent of serine-910 phosphorylated FAK (P-FAKSer910) expression in a spectrum of melanocytic proliferations as well as it's correlation with other histopathologic predictors and its effect on patient's survival. Clinicopathologic features and immunohistochemical expression of P-FAKSer910 were evaluated in 147 melanocytic proliferations: 73 primary melanoma (PM), 19 metastatic melanoma (MetM), 2 melanoma in situ, and 53 melanocytic nevi (MN). Higher cytoplasmic intensity predicted better overall survival (OS) in PM (χ=5.69; P=0.034) and was associated with 48% decrease in death risk (HR, 0.52; 95% CI, 0.28-0.95; P=0.036). In contrast, increased nuclear intensity was significantly associated with better disease-free survival (DFS) when stratified by tumor stage Log-rank test, trend of survival (χ=5.83, P=0.015) and independently on multivariate analysis when subcategorized into 3-tier categories (HR, 0.43; 95% CI, 0.18-0.98; P=0.045). Also, Clark's level and tumor-infiltrating lymphocytes (TILS) were independent predictors of DFS. Cytoplasmic intensity correlated inversely with American Joint Commission on Cancer stage in primary melanoma cases as well with vascularity in both primary and metastatic melanoma. Nuclear intensity independently correlated negatively with angioinvasion and TILS when subcategorized to 3 tier system. We found American Joint Commission on Cancer tumor stage, Clark's level, depth, ulceration, TILS, mitosis, angioinvasion, and tumor vascularity predictors of both DFS and OS. There was no significant difference in cytoplasmic or nuclear expression among the major categories of melanocytic proliferation. In this pilot immunohistochemistry-based study, we found P-FAKSer910 expression in melanoma by cytoplasmic intensity to correlate with better OS while nuclear intensity correlated with better DFS.
Collapse
|
22
|
Baier A, Szyszka R. Compounds from Natural Sources as Protein Kinase Inhibitors. Biomolecules 2020; 10:biom10111546. [PMID: 33198400 PMCID: PMC7698043 DOI: 10.3390/biom10111546] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/08/2020] [Accepted: 11/09/2020] [Indexed: 12/15/2022] Open
Abstract
The advantage of natural compounds is their lower number of side-effects when compared to most synthetic substances. Therefore, over the past several decades, the interest in naturally occurring compounds is increasing in the search for new potent drugs. Natural compounds are playing an important role as a starting point when developing new selective compounds against different diseases. Protein kinases play a huge role in several diseases, like cancers, neurodegenerative diseases, microbial infections, or inflammations. In this review, we give a comprehensive view of natural compounds, which are/were the parent compounds in the development of more potent substances using computational analysis and SAR studies.
Collapse
Affiliation(s)
- Andrea Baier
- Department of Animal Physiology and Toxicology, Institute of Biological Sciences, The John Paul II Catholic University of Lublin, 20-950 Lublin, Poland
- Correspondence:
| | - Ryszard Szyszka
- Department of Molecular Biology, Institute of Biological Sciences, The John Paul II Catholic University of Lublin, 20-950 Lublin, Poland;
| |
Collapse
|
23
|
Luo QY, Zhou SN, Pan WT, Sun J, Yang LQ, Zhang L, Qiu MZ, Yang DJ. A multi-kinase inhibitor APG-2449 enhances the antitumor effect of ibrutinib in esophageal squamous cell carcinoma via EGFR/FAK pathway inhibition. Biochem Pharmacol 2020; 183:114318. [PMID: 33159967 DOI: 10.1016/j.bcp.2020.114318] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/25/2020] [Accepted: 10/26/2020] [Indexed: 01/26/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most common types of cancer in China, with poor prognosis and lack of effective targeted therapy. It has been reported that ibrutinib possesses anticancer activity in ESCC with MYC and/or ERBB2 amplification. Here we explored the synergistic antitumor effect of a novel multi-kinase inhibitor APG-2449 with ibrutinib in ESCC and clarified the mechanism of the combination effect through in vitro and in vivo experiment. We found that APG-2449 exerted antitumor effect in ESCC. APG-2449 combined with ibrutinib showed synergistic inhibition of cell viability in ESCC cell lines. APG-2449 combined with ibrutinib dramatically inhibited the proliferation and migration of ESCC cells. Furthermore, we observed that ibrutinib combined with APG-2449 could induce more cancer cells arrested in the G1/S phase and apoptosis. In terms of mechanism, ibrutinib alone could decrease the phosphorylation level of EGFR and its downstream pathway of MEK/ERK. The combination therapy of APG-2449 and ibrutinib could significantly down-regulate the phosphorylation level of MEK/ERK and AKT. In ESCC xenotransplantation models, single therapy with either ibrutinib or APG-2449 was equivalent in delaying tumor growth, while the combination therapy suppressed tumor growth more significantly. Our data strongly suggest that the combination therapy of APG-2449 and ibrutinib can provide an effective therapeutic strategy for ESCC patients, which deserved further clinical investigation.
Collapse
Affiliation(s)
- Qiu-Yun Luo
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Su-Na Zhou
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Wen-Tao Pan
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Jian Sun
- Department of Clinical Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, PR China
| | - Li-Qiong Yang
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Lin Zhang
- Department of Clinical Laboratory, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, PR China
| | - Miao-Zhen Qiu
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, PR China.
| | - Da-Jun Yang
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China.
| |
Collapse
|
24
|
Dey A, Islam SMA, Patel R, Acevedo-Duncan M. The interruption of atypical PKC signaling and Temozolomide combination therapy against glioblastoma. Cell Signal 2020; 77:109819. [PMID: 33147518 DOI: 10.1016/j.cellsig.2020.109819] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 12/21/2022]
Abstract
Current treatment options of glioblastoma include chemotherapy and limited surgical resection. Temozolomide (TMZ) is the current therapeutic choice for chemotherapy. Still, it has severe limitations due to the development of resistance that occurs by genetic modification and constitutive activation of several cell signaling pathways. Therefore, it is essential to develop combination therapy of TMZ with other novel compounds to prevent the development of chemo-resistance. In this study, we used two inhibitors; ICA, an inhibitor of PKC-ι and ζ-Stat, an inhibitor of PKC-ζ. T98G and U87MG glioblastoma cells were treated with either ICA or ζ-stat or TMZ monotherapies, as well as TMZ were combined with either ICA or ζ-stat for five consecutive days. Our in vitro results exhibited that ICA when combined with TMZ, significantly decreased the viability of cancerous cells compared with untreated or TMZ or ICA monotherapies. Additionally, glioblastoma cells were remarkably undergoing apoptosis against the combination treatment of TMZ and ICA nucleotide compared with untreated control cells, as suggested by our Annexin-V/PI flow cytometric analysis. Moreover, the combination of TMZ and ICA also decreased the invasion of glioblastoma cell lines by acting on FAK/Paxillin pathway, as evidenced by scratch assay, transwell invasion assay, Western blot and immunoprecipitation analysis. Furthermore, our in vivo data presented that the combination of ICA and TMZ also reduced glioblastoma tumor growth and volume in mice. These data suggest that atypical PKCs, particularly PKC-ι might be an important therapeutic target as adjuvant therapy in the treatment of glioblastoma.
Collapse
Affiliation(s)
- Avijit Dey
- Department of Chemistry, University of South Florida, 4202 E Fowler Ave, Tampa, FL 33620, United States of America
| | - S M Anisul Islam
- Department of Chemistry, University of South Florida, 4202 E Fowler Ave, Tampa, FL 33620, United States of America
| | - Rekha Patel
- Department of Chemistry, University of South Florida, 4202 E Fowler Ave, Tampa, FL 33620, United States of America
| | - Mildred Acevedo-Duncan
- Department of Chemistry, University of South Florida, 4202 E Fowler Ave, Tampa, FL 33620, United States of America.
| |
Collapse
|
25
|
Li CW, Lau YT, Lam KL, Chan BP. Mechanically induced formation and maturation of 3D-matrix adhesions (3DMAs) in human mesenchymal stem cells. Biomaterials 2020; 258:120292. [DOI: 10.1016/j.biomaterials.2020.120292] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 06/15/2020] [Accepted: 08/01/2020] [Indexed: 11/26/2022]
|
26
|
P 0-Related Protein Accelerates Human Mesenchymal Stromal Cell Migration by Modulating VLA-5 Interactions with Fibronectin. Cells 2020; 9:cells9051100. [PMID: 32365526 PMCID: PMC7290418 DOI: 10.3390/cells9051100] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/16/2020] [Accepted: 04/24/2020] [Indexed: 12/22/2022] Open
Abstract
P0-related protein (PZR), a Noonan and LEOPARD syndrome target, is a member of the transmembrane Immunoglobulin superfamily. Its cytoplasmic tail contains two immune-receptor tyrosine-based inhibitory motifs (ITIMs), implicated in adhesion-dependent signaling and regulating cell adhesion and motility. PZR promotes cell migration on the extracellular matrix (ECM) molecule, fibronectin, by interacting with SHP-2 (Src homology-2 domain-containing protein tyrosine phosphatase-2), a molecule essential for skeletal development and often mutated in Noonan and LEOPARD syndrome patients sharing overlapping musculoskeletal abnormalities and cardiac defects. To further explore the role of PZR, we assessed the expression of PZR and its ITIM-less isoform, PZRb, in human bone marrow mesenchymal stromal cells (hBM MSC), and its ability to facilitate adhesion to and spreading and migration on various ECM molecules. Furthermore, using siRNA knockdown, confocal microscopy, and immunoprecipitation assays, we assessed PZR and PZRb interactions with β1 integrins. PZR was the predominant isoform in hBM MSC. Migrating hBM MSCs interacted most effectively with fibronectin and required the association of PZR, but not PZRb, with the integrin, VLA-5(α5β1), leading to modulation of focal adhesion kinase phosphorylation and vinculin levels. This raises the possibility that dysregulation of PZR function may modify hBM MSC migratory behavior, potentially contributing to skeletal abnormalities.
Collapse
|
27
|
Bennison SA, Blazejewski SM, Smith TH, Toyo-Oka K. Protein kinases: master regulators of neuritogenesis and therapeutic targets for axon regeneration. Cell Mol Life Sci 2020; 77:1511-1530. [PMID: 31659414 PMCID: PMC7166181 DOI: 10.1007/s00018-019-03336-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 09/16/2019] [Accepted: 10/08/2019] [Indexed: 12/25/2022]
Abstract
Proper neurite formation is essential for appropriate neuronal morphology to develop and defects at this early foundational stage have serious implications for overall neuronal function. Neuritogenesis is tightly regulated by various signaling mechanisms that control the timing and placement of neurite initiation, as well as the various processes necessary for neurite elongation to occur. Kinases are integral components of these regulatory pathways that control the activation and inactivation of their targets. This review provides a comprehensive summary of the kinases that are notably involved in regulating neurite formation, which is a complex process that involves cytoskeletal rearrangements, addition of plasma membrane to increase neuronal surface area, coupling of cytoskeleton/plasma membrane, metabolic regulation, and regulation of neuronal differentiation. Since kinases are key regulators of these functions during neuromorphogenesis, they have high potential for use as therapeutic targets for axon regeneration after injury or disease where neurite formation is disrupted.
Collapse
Affiliation(s)
- Sarah A Bennison
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Sara M Blazejewski
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Trevor H Smith
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Kazuhito Toyo-Oka
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA.
| |
Collapse
|
28
|
Mohanty A, Pharaon RR, Nam A, Salgia S, Kulkarni P, Massarelli E. FAK-targeted and combination therapies for the treatment of cancer: an overview of phase I and II clinical trials. Expert Opin Investig Drugs 2020; 29:399-409. [DOI: 10.1080/13543784.2020.1740680] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Atish Mohanty
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Rebecca R Pharaon
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Arin Nam
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Sabrina Salgia
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Prakash Kulkarni
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Erminia Massarelli
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
29
|
Pot M, Mihaila SM, te Brinke D, van der Borg G, Oosterwijk E, Daamen WF, van Kuppevelt TH. Introduction of Specific 3D Micromorphologies in Collagen Scaffolds Using Odd and Even Dicarboxylic Acids. ACS OMEGA 2020; 5:3908-3916. [PMID: 32149217 PMCID: PMC7057318 DOI: 10.1021/acsomega.9b03350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 02/10/2020] [Indexed: 05/03/2023]
Abstract
The construction of scaffolds and subsequent incorporation of cells and biologics have been widely investigated to regenerate damaged tissues. Scaffolds act as a template to guide tissue formation, and their characteristics have a considerable impact on the regenerative process. Whereas many technologies exist to induce specific two-dimensional (2D) morphologies into biomaterials, the introduction of three-dimensional (3D) micromorphologies into individual pore walls of scaffolds produced from biological molecules such as collagen poses a challenge. We here report the use of dicarboxylic acids to induce specific micromorphologies in collagen scaffolds and evaluate their effect on cellular migration and differentiation. Insoluble type I collagen fibrils were suspended in monocarboxylic and dicarboxylic acids of different concentrations, and unidirectional and random pore scaffolds were constructed by freezing and lyophilization. The application of various acids and concentrations resulted in variations in 3D micromorphologies, including wall structure, wall thickness, and pore size. The use of dicarboxylic acids resulted in acid-specific micromorphologies, whereas monocarboxylic acids did not. Dicarboxylic acids with an odd or even number of C-atoms resulted in frayed/fibrillar or smooth wall structures, respectively, with varying appearances. The formation of micromorphologies was concentration-dependent. In vitro analysis indicated the cytocompatibility of scaffolds, and micromorphology-related cell behavior was indicated by enhanced myosin staining and myosin heavy chain gene expression for C2C12 myoblasts cultured on scaffolds with frayedlike micromorphologies compared to those with smooth micromorphologies. In conclusion, porous collagen scaffolds with various intrawall 3D micromorphologies can be constructed by application of dicarboxylic acids, superimposing the second level of morphology to the overall scaffold structure. Acid crystal formation is key to the specific micromorphologies observed and can be explained by the odd/even theory for dicarboxylic acids. Scaffolds with a 3D micrometer-defined topography may be used as a screening platform to select optimal substrates for the regeneration of specific tissues.
Collapse
Affiliation(s)
- Michiel
W. Pot
- Department
of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Silvia M. Mihaila
- Department
of Urology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Dana te Brinke
- Department
of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Guus van der Borg
- Department
of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Egbert Oosterwijk
- Department
of Urology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Willeke F. Daamen
- Department
of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Toin H. van Kuppevelt
- Department
of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| |
Collapse
|
30
|
Perri AM, Agosti V, Olivo E, Concolino A, Angelis MD, Tammè L, Fiumara CV, Cuda G, Scumaci D. Histone proteomics reveals novel post-translational modifications in breast cancer. Aging (Albany NY) 2019; 11:11722-11755. [PMID: 31816600 PMCID: PMC6932915 DOI: 10.18632/aging.102577] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 11/26/2019] [Indexed: 04/28/2023]
Abstract
Histones and their variants are subjected to several post-translational modifications (PTMs). Histones PTMs play an important role in the regulation of gene expression and are critical for the development and progression of many types of cancer, including breast cancer. In this study, we used two-dimensional TAU/SDS electrophoresis, coupled with mass spectrometry for a comprehensive profiling of histone PTMs in breast cancer cell lines.Proteomic approach allowed us to identify 85 histone PTMs, seventeen of which are not reported in the UniProt database. Western blot analysis was performed to confirm a peculiar pattern of PTMs in the sporadic and hereditary breast cancer cell lines compared to normal cells. Overlapping mass spectrometry data with western blotting results, we identified, for the first time to our knowledge, a tyrosine phosphorylation on histone H1, which is significantly higher in breast cancer cells. Additionally, by inhibiting specific signaling paths, such as PI3K, PPARγ and FAK pathways, we established a correlation between their regulation and the presence of new histone PTMs. Our results may provide new insight on the possible implication of these modifications in breast cancer and may offer new perspectives for future clinical applications.
Collapse
Affiliation(s)
- Angela Mena Perri
- Laboratory of Proteomics, Research Center on Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Salvatore Venuta University Campus, Catanzaro, Italy
| | - Valter Agosti
- Laboratory of Molecular Oncology, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy, CIS for Genomics and Molecular Pathology, Magna Graecia University, Catanzaro, Italy
| | - Erika Olivo
- Laboratory of Proteomics, Research Center on Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Salvatore Venuta University Campus, Catanzaro, Italy
| | - Antonio Concolino
- Laboratory of Proteomics, Research Center on Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Salvatore Venuta University Campus, Catanzaro, Italy
| | - MariaTeresa De Angelis
- Stem Cell Laboratory, Research Center of Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine University “Magna Graecia” of Catanzaro, Salvatore Venuta University Campus, Catanzaro, Italy
| | - Laura Tammè
- Laboratory of Proteomics, Research Center on Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Salvatore Venuta University Campus, Catanzaro, Italy
| | - Claudia Vincenza Fiumara
- Laboratory of Proteomics, Research Center on Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Salvatore Venuta University Campus, Catanzaro, Italy
| | - Giovanni Cuda
- Laboratory of Proteomics, Research Center on Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Salvatore Venuta University Campus, Catanzaro, Italy
| | - Domenica Scumaci
- Laboratory of Proteomics, Research Center on Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Salvatore Venuta University Campus, Catanzaro, Italy
| |
Collapse
|
31
|
Deville SS, Cordes N. The Extracellular, Cellular, and Nuclear Stiffness, a Trinity in the Cancer Resistome-A Review. Front Oncol 2019; 9:1376. [PMID: 31867279 PMCID: PMC6908495 DOI: 10.3389/fonc.2019.01376] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/22/2019] [Indexed: 12/19/2022] Open
Abstract
Alterations in mechano-physiological properties of a tissue instigate cancer burdens in parallel to common genetic and epigenetic alterations. The chronological and mechanistic interrelation between the various extra- and intracellular aspects remains largely elusive. Mechano-physiologically, integrins and other cell adhesion molecules present the main mediators for transferring and distributing forces between cells and the extracellular matrix (ECM). These cues are channeled via focal adhesion proteins, termed the focal adhesomes, to cytoskeleton and nucleus and vice versa thereby affecting the pathophysiology of multicellular cancer tissues. In combination with simultaneous activation of diverse downstream signaling pathways, the phenotypes of cancer cells are created and driven characterized by deregulated transcriptional and biochemical cues that elicit the hallmarks of cancer. It, however, remains unclear how elastostatic modifications, i.e., stiffness, in the extracellular, intracellular, and nuclear compartment contribute and control the resistance of cancer cells to therapy. In this review, we discuss how stiffness of unique tumor components dictates therapy response and what is known about the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Sara Sofia Deville
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Helmholtz-Zentrum Dresden - Rossendorf, Technische Universität Dresden, Dresden, Germany
- Department of Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Nils Cordes
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Helmholtz-Zentrum Dresden - Rossendorf, Technische Universität Dresden, Dresden, Germany
- Department of Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
- German Cancer Consortium (DKTK), Dresden, Germany
- Germany German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
32
|
Aboubakar Nana F, Hoton D, Ambroise J, Lecocq M, Vanderputten M, Sibille Y, Vanaudenaerde B, Pilette C, Bouzin C, Ocak S. Increased Expression and Activation of FAK in Small-Cell Lung Cancer Compared to Non-Small-Cell Lung Cancer. Cancers (Basel) 2019; 11:cancers11101526. [PMID: 31658694 PMCID: PMC6827365 DOI: 10.3390/cancers11101526] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 10/02/2019] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Focal adhesion kinase (FAK) plays a crucial role in cancer development and progression. FAK is overexpressed and/or activated and associated with poor prognosis in various malignancies. However, in lung cancer, activated FAK expression and its prognostic value are unknown. METHODS FAK and activated FAK (phospho-FAK Y397) expressions were analyzed by multiplex immunofluorescence staining in formalin-fixed paraffin-embedded tissues from 95 non-small-cell lung cancer (NSCLC) and 105 small-cell lung cancer (SCLC) patients, and 37 healthy donors. The FAK staining score was defined as the percentage (%) of FAK-stained tumor area multiplied by (×) FAK mean intensity and phospho-FAK staining score as the (% of phospho-FAK-stained area of low intensity × 1) + (% of phospho-FAK-stained area of medium intensity × 2) + (% of the phospho-FAK-stained area of high intensity × 3). FAK and phospho-FAK staining scores were compared between normal, NSCLC, and SCLC tissues. They were also tested for correlations with patient characteristics and clinical outcomes. RESULTS The median follow-up time after the first treatment was 42.5 months and 6.4 months for NSCLC and SCLC patients, respectively. FAK and phospho-FAK staining scores were significantly higher in lung cancer than in normal lung and significantly higher in SCLC compared to NSCLC tissues (p < 0.01). Moreover, the ratio between phospho-FAK and FAK staining scores was significantly higher in SCLC than in NSCLC tissues (p < 0.01). However, FAK and activated FAK expression in lung cancer did not correlate with recurrence-free and overall survival in NSCLC and SCLC patients. CONCLUSIONS Total FAK and activated FAK expressions are significantly higher in lung cancer than in normal lung, and significantly higher in SCLC compared to NSCLC, but are not prognostic biomarkers in this study.
Collapse
Affiliation(s)
- Frank Aboubakar Nana
- Pole of Pneumology, ENT, and Dermatology (PNEU), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
- Division of Pneumology, Cliniques Universitaires St-Luc, UCLouvain, 1200 Brussels, Belgium.
| | - Delphine Hoton
- Department of Pathology, Cliniques Universitaires Saint-Luc, UCLouvain, 1200 Brussels, Belgium.
| | - Jérôme Ambroise
- Centre de Technologies Moléculaires Appliquées, IREC, UCLouvain, 1200 Brussels, Belgium.
| | - Marylène Lecocq
- Pole of Pneumology, ENT, and Dermatology (PNEU), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
| | - Marie Vanderputten
- Pole of Pneumology, ENT, and Dermatology (PNEU), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
| | - Yves Sibille
- Pole of Pneumology, ENT, and Dermatology (PNEU), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
- Division of Pneumology, CHU UCL Namur (Godinne Site), UCLouvain, 5530 Yvoir, Belgium.
| | - Bart Vanaudenaerde
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Belgium.
| | - Charles Pilette
- Pole of Pneumology, ENT, and Dermatology (PNEU), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
- Division of Pneumology, Cliniques Universitaires St-Luc, UCLouvain, 1200 Brussels, Belgium.
| | | | - Sebahat Ocak
- Pole of Pneumology, ENT, and Dermatology (PNEU), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
- Division of Pneumology, CHU UCL Namur (Godinne Site), UCLouvain, 5530 Yvoir, Belgium.
| |
Collapse
|
33
|
Rajshankar D, Wang B, Worndl E, Menezes S, Wang Y, McCulloch CA. Focal adhesion kinase regulates tractional collagen remodeling, matrix metalloproteinase expression, and collagen structure, which in turn affects matrix‐induced signaling. J Cell Physiol 2019; 235:3096-3111. [DOI: 10.1002/jcp.29215] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 09/03/2019] [Indexed: 11/08/2022]
Affiliation(s)
| | - Baiyu Wang
- Faculty of Dentistry University of Toronto Toronto Ontario
| | | | - Sara Menezes
- Faculty of Dentistry University of Toronto Toronto Ontario
| | - Yongqiang Wang
- Faculty of Dentistry University of Toronto Toronto Ontario
| | | |
Collapse
|
34
|
Osipov A, Saung MT, Zheng L, Murphy AG. Small molecule immunomodulation: the tumor microenvironment and overcoming immune escape. J Immunother Cancer 2019; 7:224. [PMID: 31439034 PMCID: PMC6704558 DOI: 10.1186/s40425-019-0667-0] [Citation(s) in RCA: 160] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 07/08/2019] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy has led to a paradigm shift in the treatment of many advanced malignancies. Despite the success in treatment of tumors like non-small cell lung cancer (NSCLC) and melanoma, checkpoint inhibition-based immunotherapy has limitations. Many tumors, such as pancreatic cancer, are less responsive to checkpoint inhibitors, where patients tend to have a limited duration of benefit and where clinical responses are more robust in patients who are positive for predictive biomarkers. One of the critical factors that influence the efficacy of immunotherapy is the tumor microenvironment (TME), which contains a heterogeneous composition of immunosuppressive cells. Myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) alter the immune landscape of the TME and serve as facilitators of tumor proliferation, metastatic growth and immunotherapy resistance. Small molecule inhibitors that target these components of the TME have been developed. This special issue review focuses on two promising classes of immunomodulatory small molecule inhibitors: colony stimulating factor-1 receptor (CSF-1R) and focal adhesion kinase (FAK). Small molecule inhibitors of CSF-1R reprogram the TME and TAMs, and lead to enhanced T-cell-mediated tumor eradication. FAK small molecule inhibitors decrease the infiltration MDSCs, TAMs and regulatory T-cells. Additionally, FAK inhibitors are implicated as modulators of stromal density and cancer stem cells, leading to a TME more conducive to an anti-tumor immune response. Immunomodulatory small molecule inhibitors present a unique opportunity to attenuate immune escape of tumors and potentiate the effectiveness of immunotherapy and traditional cytotoxic therapy.
Collapse
Affiliation(s)
- Arsen Osipov
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - May Tun Saung
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lei Zheng
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Adrian G Murphy
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- GI Oncology, Sidney Kimmel Comprehensive Cancer Center, Harry and Jeanette Weinberg Building, CRB1 1, Room 487, 1650 Orleans Street, Baltimore, MD, 21231, USA.
| |
Collapse
|
35
|
Zhou J, Yi Q, Tang L. The roles of nuclear focal adhesion kinase (FAK) on Cancer: a focused review. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:250. [PMID: 31186061 PMCID: PMC6560741 DOI: 10.1186/s13046-019-1265-1] [Citation(s) in RCA: 201] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 06/03/2019] [Indexed: 12/15/2022]
Abstract
FAK is a tyrosine kinase overexpressed in cancer cells and plays an important role in the progression of tumors to a malignant phenotype. Except for its typical role as a cytoplasmic kinase downstream of integrin and growth factor receptor signaling, related studies have shown new aspects of the roles of FAK in the nucleus. FAK can promote p53 degradation through ubiquitination, leading to cancer cell growth and proliferation. FAK can also regulate GATA4 and IL-33 expression, resulting in reduced inflammatory responses and immune escape. These findings establish a new model of FAK from the cytoplasm to the nucleus. Activated FAK binds to transcription factors and regulates gene expression. Inactive FAK synergizes with different E3 ligases to promote the turnover of transcription factors by enhancing ubiquitination. In the tumor microenvironment, nuclear FAK can regulate the formation of new blood vessels, affecting the tumor blood supply. This article reviews the roles of nuclear FAK in regulating gene expression. In addition, the use of FAK inhibitors to target nuclear FAK functions will also be emphasized.
Collapse
Affiliation(s)
- Jin Zhou
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Qian Yi
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Liling Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
36
|
Andisha NM, McMillan DC, Gujam FJA, Roseweir A, Edwards J. The relationship between phosphorylation status of focal adhesion kinases, molecular subtypes, tumour microenvironment and survival in patients with primary operable ductal breast cancer. Cell Signal 2019; 60:91-99. [PMID: 30981841 DOI: 10.1016/j.cellsig.2019.04.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/01/2019] [Accepted: 04/10/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Despite advances in therapies to treat breast cancer, over 100,000 patients die in the UK of this disease per year, highlighting the need to develop effect predictive and prognostic markers for patients with primary operable ductal breast cancer. Therefore, the aim of the present study was to examine the relationship between membranous, cytoplasmic and nuclear expression of focal adhesion kinase (phosphorylated at Y 397, Y 861 and Y 925), molecular subtypes, tumour microenvironment and survival in patients with primary operable ductal breast cancer. METHODS Four hundred and seventy-four patients presenting between 1995 and 1998 with primary operable ductal breast cancer were included in this study. Using tissue microarrays expression of membranous, cytoplasmic and nuclear tumour cell phosphorylation of FAK at Y397, Y861 and Y925 was assessed, and associations with clinicopathological characteristics, tumour microenvironment and cancer-specific survival (CSS) were examined. RESULTS No significant association was observed for ph-FAK Y861 with survival at all sites. However, high expression of membranous ph-FAK Y397 was associated with increased tumour grade (P < .001), molecular subtypes (P < .001), increased tumour necrosis (P < .001), high Klintrup-Mäkinen grade (P < .001), increased CD138+ plasma cells (P = .031), endocrine therapy (P = .001) and poor cancer specific survival (P = .040). Similarly, high expression of nuclear ph-FAK Y397 was associated with decreased age (P = .042), increased CD138+ plasma cells (P = .001) and poor cancer specific survival (P = .003). Furthermore, high expression of cytoplasmic ph-FAK Y925 was associated with decreased tumour grade (P < .001), less involved lymph node (P = .020), molecular subtypes (P < .001), decreased tumour necrosis (P < .001), low Klintrup-Mäkinen grade (P < .001), decreased CD4+ T-cells (P = .006), decreased CD138+ plasma cells (P = .034), endocrine therapy (P < .001), chemotherapy (P = .048), and improved cancer specific survival (P = .044). On multivariate analysis, high expression of nuclear ph-FAK Y397 was independently associated with reduced cancer specific survival (P = .017). CONCLUSION The results of the present study show that membranous and nuclear ph-FAK Y397 and cytoplasmic ph-FAK Y925 were associated with prognosis in patients with primary operable ductal breast cancer. In addition, high expression of nuclear ph-FAK Y397 was an independent prognostic factor in patients with primary operable ductal breast cancer and could be incorporated into clinical practice.
Collapse
Affiliation(s)
- Najla M Andisha
- Academic Unit of Surgery, College of Medical, Veterinary and Life Sciences-University of Glasgow, Royal Infirmary, Glasgow, UK; Unit of Gastrointestinal cancer and Molecular Pathology, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences-University of Glasgow, Glasgow, UK.
| | - Donald C McMillan
- Academic Unit of Surgery, College of Medical, Veterinary and Life Sciences-University of Glasgow, Royal Infirmary, Glasgow, UK
| | - Fadia J A Gujam
- Academic Unit of Surgery, College of Medical, Veterinary and Life Sciences-University of Glasgow, Royal Infirmary, Glasgow, UK; Unit of Gastrointestinal cancer and Molecular Pathology, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences-University of Glasgow, Glasgow, UK
| | - Antonia Roseweir
- Unit of Gastrointestinal cancer and Molecular Pathology, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences-University of Glasgow, Glasgow, UK
| | - Joanne Edwards
- Unit of Gastrointestinal cancer and Molecular Pathology, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences-University of Glasgow, Glasgow, UK
| |
Collapse
|
37
|
Increased expression of FAK isoforms as potential cancer biomarkers in ovarian cancer. Oncol Lett 2019; 17:4779-4786. [PMID: 31186683 PMCID: PMC6507456 DOI: 10.3892/ol.2019.10147] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 12/07/2018] [Indexed: 12/20/2022] Open
Abstract
Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase that is expressed in most human cell types (example: Epithelial cells, fibroblasts and endothelial), it serves a key role in the control of cell survival, proliferation and motility. The abnormal expression of FAK has been associated with poor prognosis in cancer, including ovarian cancer. However, although FAK isoforms with specific molecular and functional properties have been characterized, there are a limited number of published studies that examine FAK isoforms in ovarian cancer. The aim of the present study was to analyze the expression level of FAK and its isoforms in ovarian cancer. The expression of FAK kinase and focal adhesion targeting (FAT) domains was determined with immunohistochemistry in healthy ovary, and serous and mucinous cystadenoma, borderline tumor and carcinoma samples. Additionally, the expression of FAK and its isoforms were investigated in three ovarian cancer-derived cell lines with western blotting and reverse transcription-semi-quantitative polymerase chain reaction. An increased expression of FAK kinase domain was determined in serous tumor samples and was associated with advancement of the lesion. FAK kinase domain expression was moderate-to-low in mucinous tumor samples. The expression of the FAK FAT domain in tumor samples was reduced, compared with healthy ovary samples; however, the FAT domain was localized to the cellular nucleus. Expression of alternative transcripts FAK°, FAK28,6 and FAK28 was determined in all three cell lines investigated. In conclusion, FAK kinase and FAT domains are differentially expressed among ovarian tumor types. These results indicated the presence of at least two isoforms of FAK (FAK and the putative FAK-related non-kinase) in tumor tissue, which is supported by the cells producing at least three FAK alternative transcripts. These results may support the use of FAK and its isoforms as biomarkers for ovarian cancer.
Collapse
|
38
|
Chuang HH, Wang PH, Niu SW, Zhen YY, Huang MS, Hsiao M, Yang CJ. Inhibition of FAK Signaling Elicits Lamin A/C-Associated Nuclear Deformity and Cellular Senescence. Front Oncol 2019; 9:22. [PMID: 30761269 PMCID: PMC6363943 DOI: 10.3389/fonc.2019.00022] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 01/08/2019] [Indexed: 01/07/2023] Open
Abstract
Focal adhesion kinase (FAK) is a non-receptor kinase that facilitates tumor aggressiveness. The effects of FAK inhibition include arresting proliferation, limiting metastasis, and inhibiting angiogenesis. PF-573228 is an ATP-competitive inhibitor of FAK. Treating lung cancer cells with PF-573228 resulted in FAK inactivation and changes in the expressions of lamin A/C and nuclear deformity. Since lamin A/C downregulation or deficiency was associated with cellular senescence, the senescence-associated β-galactosidase (SA-β-gal) assay was used to investigate whether PF-573228 treatment drove cellular senescence, which showed more SA-β-gal-positive cells in culture. p53 is known to play a pivotal role in mediating the progression of cellular senescence, and the PF-573228-treated lung cancer cells resulted in a higher p53 expression level. Subsequently, the FAK depletion in lung cancer cells was employed to confirm the role of FAK inhibition on cellular senescence. FAK depletion and pharmacological inhibition of lung cancer cells elicited similar patterns of cellular senescence, lamin A/C downregulation, and p53 upregulation, implying that FAK signaling is associated with the expression of p53 and the maintenance of lamin A/C levels to shape regular nuclear morphology and manage anti-senescence. Conversely, FAK inactivation led to p53 upregulation, disorganization of the nuclear matrix, and consequently cellular senescence. Our data suggest a new FAK signaling pathway, in that abolishing FAK signaling can activate the senescence program in cells. Triggering cellular senescence could be a new therapeutic approach to limit tumor growth.
Collapse
Affiliation(s)
- Hsiang-Hao Chuang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Pei-Hui Wang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Sheng-Wen Niu
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yen-Yi Zhen
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Shyan Huang
- Department of Internal Medicine, E-Da Cancer Hospital, School of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.,Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Jen Yang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
39
|
Smyrek I, Mathew B, Fischer SC, Lissek SM, Becker S, Stelzer EHK. E-cadherin, actin, microtubules and FAK dominate different spheroid formation phases and important elements of tissue integrity. Biol Open 2019; 8:bio.037051. [PMID: 30578251 PMCID: PMC6361217 DOI: 10.1242/bio.037051] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Spheroids resemble features of tissues and serve as model systems to study cell–cell and cell–ECM interactions in non-adhesive three-dimensional environments. Although it is generally accepted that mature spheroids resemble tissue properties very well, no studies relate different phases in the spheroid formation processes that contribute to tissue integrity. Tissue integrity involves the cellular processes adhesion formation, adhesion reinforcement, rearrangement as well as proliferation. They maintain the structure and function of tissues and, upon dysregulation, contribute to malignancy. We investigated spheroid formation dynamics in cell lines of different metastatic potential. We dissected spheroid formation into phases of aggregation, compaction and growth to identify the respective contributions of E-cadherin, actin, microtubules and FAK. E-cadherin, actin and microtubules drive the first two phases. Microtubules and FAK are involved in the proliferation phase. FAK activity correlates with the metastatic potential of the cells. A robust computational model based on a very large number of experiments reveals the temporal resolution of cell adhesion. Our results provide novel hypotheses to unveil the general mechanisms that contribute to tissue integrity. Summary: The phases of spheroid formation resemble different stages of cell contact formation. This facilitates studying the temporal contribution of molecules in this process.
Collapse
Affiliation(s)
- I Smyrek
- Physical Biology/Physikalische Biologie (IZN, FB 15), Buchmann Institute for Molecular Life Sciences (BMLS), Cluster of Excellence Frankfurt - Macromolecular Complexes (CEF - MC), Goethe Universität - Frankfurt am Main (Campus Riedberg), Max-von-Laue-Straße 15, D-60348 Frankfurt am Main, Germany
| | - B Mathew
- Physical Biology/Physikalische Biologie (IZN, FB 15), Buchmann Institute for Molecular Life Sciences (BMLS), Cluster of Excellence Frankfurt - Macromolecular Complexes (CEF - MC), Goethe Universität - Frankfurt am Main (Campus Riedberg), Max-von-Laue-Straße 15, D-60348 Frankfurt am Main, Germany
| | - S C Fischer
- Physical Biology/Physikalische Biologie (IZN, FB 15), Buchmann Institute for Molecular Life Sciences (BMLS), Cluster of Excellence Frankfurt - Macromolecular Complexes (CEF - MC), Goethe Universität - Frankfurt am Main (Campus Riedberg), Max-von-Laue-Straße 15, D-60348 Frankfurt am Main, Germany
| | - S M Lissek
- Physical Biology/Physikalische Biologie (IZN, FB 15), Buchmann Institute for Molecular Life Sciences (BMLS), Cluster of Excellence Frankfurt - Macromolecular Complexes (CEF - MC), Goethe Universität - Frankfurt am Main (Campus Riedberg), Max-von-Laue-Straße 15, D-60348 Frankfurt am Main, Germany
| | - S Becker
- Physical Biology/Physikalische Biologie (IZN, FB 15), Buchmann Institute for Molecular Life Sciences (BMLS), Cluster of Excellence Frankfurt - Macromolecular Complexes (CEF - MC), Goethe Universität - Frankfurt am Main (Campus Riedberg), Max-von-Laue-Straße 15, D-60348 Frankfurt am Main, Germany
| | - E H K Stelzer
- Physical Biology/Physikalische Biologie (IZN, FB 15), Buchmann Institute for Molecular Life Sciences (BMLS), Cluster of Excellence Frankfurt - Macromolecular Complexes (CEF - MC), Goethe Universität - Frankfurt am Main (Campus Riedberg), Max-von-Laue-Straße 15, D-60348 Frankfurt am Main, Germany
| |
Collapse
|
40
|
Joshi H, Morley SC. Cells under stress: The mechanical environment shapes inflammasome responses to danger signals. J Leukoc Biol 2019; 106:119-125. [PMID: 30645000 DOI: 10.1002/jlb.3mir1118-417r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 12/19/2018] [Accepted: 12/22/2018] [Indexed: 12/11/2022] Open
Abstract
Many intracellular signals, such as host danger-associated molecules and bacterial toxins during infection, elicit inflammasome activation. However, the mechanical environment in tissues may also influence the sensitivity of various inflammasomes to activation. The cellular mechanical environment is determined by the extracellular tissue stiffness, or its inverse, tissue compliance. Tissue stiffness is sensed by the intracellular cytoskeleton through a process termed mechanotransduction. Thus, extracellular compliance and the intracellular cytoskeleton may regulate the sensitivity of inflammasome activation. Control of proinflammatory signaling by tissue compliance may contribute to the pathogenesis of diseases such as ventilator-induced lung injury during bacterial pneumonia and tissue fibrosis in inflammatory disorders. The responsible signaling cascades in inflammasome activation pathways and mechanotransduction crosstalk are not yet fully understood. This rather different immunomodulatory perspective will be reviewed and open questions discussed here.
Collapse
Affiliation(s)
- Hemant Joshi
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA.,Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sharon Celeste Morley
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA.,Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
41
|
Mechanosensing in liver regeneration. Semin Cell Dev Biol 2017; 71:153-167. [DOI: 10.1016/j.semcdb.2017.07.041] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 12/13/2022]
|
42
|
Canel M, Byron A, Sims AH, Cartier J, Patel H, Frame MC, Brunton VG, Serrels B, Serrels A. Nuclear FAK and Runx1 Cooperate to Regulate IGFBP3, Cell-Cycle Progression, and Tumor Growth. Cancer Res 2017; 77:5301-5312. [PMID: 28807942 PMCID: PMC6126615 DOI: 10.1158/0008-5472.can-17-0418] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 06/23/2017] [Accepted: 08/04/2017] [Indexed: 12/20/2022]
Abstract
Nuclear focal adhesion kinase (FAK) is a potentially important regulator of gene expression in cancer, impacting both cellular function and the composition of the surrounding tumor microenvironment. Here, we report in a murine model of skin squamous cell carcinoma (SCC) that nuclear FAK regulates Runx1-dependent transcription of insulin-like growth factor binding protein 3 (IGFBP3), and that this regulates SCC cell-cycle progression and tumor growth in vivo Furthermore, we identified a novel molecular complex between FAK and Runx1 in the nucleus of SCC cells and showed that FAK interacted with a number of Runx1-regulatory proteins, including Sin3a and other epigenetic modifiers known to alter Runx1 transcriptional function through posttranslational modification. These findings provide important new insights into the role of FAK as a scaffolding protein in molecular complexes that regulate gene transcription. Cancer Res; 77(19); 5301-12. ©2017 AACR.
Collapse
Affiliation(s)
- Marta Canel
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Adam Byron
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew H Sims
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Jessy Cartier
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, United Kingdom
| | - Hitesh Patel
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Margaret C Frame
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Valerie G Brunton
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Bryan Serrels
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom.
| | - Alan Serrels
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom.
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
43
|
Buhrmann C, Shayan P, Goel A, Shakibaei M. Resveratrol Regulates Colorectal Cancer Cell Invasion by Modulation of Focal Adhesion Molecules. Nutrients 2017; 9:E1073. [PMID: 28953264 PMCID: PMC5691690 DOI: 10.3390/nu9101073] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 09/08/2017] [Accepted: 09/18/2017] [Indexed: 01/19/2023] Open
Abstract
Resveratrol, a safe and multi-targeted agent, has been associated with suppression of survival, proliferation and metastasis of cancer, however, the underlying mechanisms for its anti-cancer activity, particularly on cellular signaling during cancer cell migration still remain poorly understood. We investigated the invasion response of two human colorectal cancer (CRC) cells (HCT116 and SW480) to resveratrol and studied the effect of specific pharmacological inhibitors, cytochalasin D (CytD) and focal adhesion kinase-inhibitor (FAK-I) on FAK, cell viability and migration in CRC. We found that resveratrol altered cell phenotype of both CRC cells, reduced cell viability and the results were comparable to FAK-I and CytD. These effects of resveratrol were associated with marked Sirt1 up-regulation, FAK down-regulation, inhibition of focal adhesion and potentiation of effects by combinatorial treatment of resveratrol and inhibitors. Interestingly, inhibition of FAK with FAK-I or treatment with CytD suppressed resveratrol-induced Sirt1 up-regulation and markedly down-regulated FAK expression. Resveratrol or combination treatment with inhibitors significantly activated caspase-3 and potentiated apoptosis. Moreover, resveratrol suppressed invasion and colony forming capacity, cell proliferation, β1-Integrin expression and activation of FAK of cells in alginate tumor microenvironment, similar to FAK-I or CytD. Finally, we demonstrated that resveratrol, FAK-I or CytD inhibited activation of NF-κB, suppressed NF-κB-dependent gene end-products involved in invasion, metastasis, and apoptosis; and these effects of resveratrol were potentiated by combination treatment with FAK-I or CytD. Our data illustrated that the anti-invasion effect of resveratrol by inhibition of FAK activity has a potential beneficial role in disease prevention and therapeutic management of CRC.
Collapse
Affiliation(s)
- Constanze Buhrmann
- Musculoskeletal Research Group and Tumour Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Pettenkoferstrasse 11, D-80336 Munich, Germany.
| | - Parviz Shayan
- Department of Parasitology, Faculty of Veterinary Medicine, University of Tehran, Tehran 141556453, Iran.
- Investigating Institute of Molecular Biological System Transfer, Tehran 1417863171, Iran.
| | - Ajay Goel
- Center for Gastrointestinal Research, Center for Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A Sammons Cancer Center, Baylor University Medical Center, Dallas, TX 75246, USA.
| | - Mehdi Shakibaei
- Musculoskeletal Research Group and Tumour Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Pettenkoferstrasse 11, D-80336 Munich, Germany.
| |
Collapse
|
44
|
Damayanti NP, Buno K, Narayanan N, Voytik Harbin SL, Deng M, Irudayaraj JMK. Monitoring focal adhesion kinase phosphorylation dynamics in live cells. Analyst 2017; 142:2713-2716. [PMID: 28589989 PMCID: PMC5531600 DOI: 10.1039/c7an00471k] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Focal adhesion kinase (FAK) is a cytoplasmic non-receptor tyrosine kinase essential for a diverse set of cellular functions. Current methods for monitoring FAK activity in response to an extracellular stimulus lack spatiotemporal resolution and/or the ability to perform multiplex detection. Here we report on a novel approach to monitor the real-time kinase phosphorylation activity of FAK in live single cells by fluorescence lifetime imaging.
Collapse
Affiliation(s)
- Nur P Damayanti
- Bindley Bioscience Center, Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, USA. and Indiana University School of Medicine, 980 West Walnut St. R3 C312, Indianapolis, Indiana 46202, USA
| | - Kevin Buno
- Weldon School of Biomedical Engineering, Collage of Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| | - Nagarajan Narayanan
- Bindley Bioscience Center, Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, USA.
| | - Sherry L Voytik Harbin
- Weldon School of Biomedical Engineering, Collage of Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| | - Meng Deng
- Bindley Bioscience Center, Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, USA.
| | - Joseph M K Irudayaraj
- Bindley Bioscience Center, Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, USA.
| |
Collapse
|
45
|
Patil S, Sarode GS, Sarode SC, Anand R, Patil S. Focal Adhesion Kinase Expression in Ameloblastoma: A Preliminary Observational Study. J Clin Diagn Res 2017; 11:ZC47-ZC50. [PMID: 28764292 DOI: 10.7860/jcdr/2017/28895.10064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/05/2017] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Focal Adhesion Kinase (FAK) is a membrane-associated molecule, which plays a major role in tumourigenesis by promoting cell survival, proliferation, migration, and invasion. Hence, FAK is a well known molecule that has been targeted therapeutically for the treatment of various malignancies. Ameloblastoma (AB) is a locally invasive, destructive and recurrent benign odontogenic lesion. Based on these properties, we have hypothesized that odontogenic epithelium of AB could show increased expression of FAK. AIM To study the expression of FAK in odontogenic epithelium of AB using immunohistochemistry. MATERIALS AND METHODS Thirty four paraffin embedded AB and 17 Dental Follicle (DF) tissues were retrieved from archives. Immunohistochemical staining was performed using FAK antibody on 5 μm thick formalin fixed paraffin embedded sections. The expression of FAK was studied in ameloblast like cells and stellate reticulum-like cells and was graded as negative, weak and strong. The data was analysed using chi-square test and Fisher's-exact test. RESULTS In AB, both ameloblast like and stellate reticulum like cells showed positive expression of FAK in the cytoplasm in all the cases (34). The epithelial islands from all the DFs (17) were negative for FAK expression. The difference in the expression of FAK in ameloblast like and stellate reticulum like cells was statistically significant (p<0.0001). Statistically significant difference was observed in the expression of FAK in stellate reticulum like cells of AB and odontogenic epithelial islands of DF (p = 0.0017). All the AB cases showed strong expression of FAK in ameloblast like cells and negative expression was seen in all the cases of DF. CONCLUSION FAK was strongly expressed in AB as compared to epithelial islands of DF. FAK could be responsible for the neoplastic nature of AB and possibly play a role in tumour invasiveness.
Collapse
Affiliation(s)
- Snehal Patil
- Postgraduate Student, Department of Oral Pathology and Microbiology, Dr. D. Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Sant-Tukaram Nagar, Pimpri, Pune, Maharashtra, India
| | - Gargi Sachin Sarode
- Associate Professor, Department of Oral Pathology and Microbiology, Dr. D. Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Sant-Tukaram Nagar, Pimpri, Pune, Maharashtra, India
| | - Sachin C Sarode
- Professor, Department of Oral Pathology and Microbiology, Dr. D. Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Sant-Tukaram Nagar, Pimpri, Pune, Maharashtra, India
| | - Rahul Anand
- Postgraduate Student, Department of Oral Pathology and Microbiology, Dr. D. Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Sant-Tukaram Nagar, Pimpri, Pune, Maharashtra, India
| | - Shankargouda Patil
- Associate Professor, Department of Oral and Maxillofacial Surgery, Division of Oral Pathology, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| |
Collapse
|
46
|
E-cadherin expression is correlated with focal adhesion kinase inhibitor resistance in Merlin-negative malignant mesothelioma cells. Oncogene 2017; 36:5522-5531. [PMID: 28553954 DOI: 10.1038/onc.2017.147] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 03/13/2017] [Accepted: 04/14/2017] [Indexed: 12/15/2022]
Abstract
Malignant mesothelioma (MM) is an aggressive tumor commonly caused by asbestos exposure after a long latency. Focal adhesion kinase (FAK) inhibitors inhibit the cell growth of Merlin-deficient MM cells; however, their clinical efficacy has not been clearly determined. The aim of this study was to evaluate the growth inhibitory effect of the FAK inhibitor VS-4718 on MM cell lines and identify biomarkers for its efficacy. Although most Merlin-deficient cell lines were sensitive to VS-4718 compared with control MeT-5A cells, a subset of these cell lines exhibited resistance to this drug. Microarray and qRT-PCR analyses using RNA isolated from Merlin-deficient MM cell lines revealed a significant correlation between E-cadherin mRNA levels and VS-4718 resistance. Merlin- and E-cadherin-negative Y-MESO-22 cells underwent apoptosis upon treatment with a low concentration of VS-4718, whereas Merlin-negative, E-cadherin-positive Y-MESO-9 cells did not undergo VS-4718-induced apoptosis. Furthermore, E-cadherin knockdown in Merlin-negative MM cells significantly sensitized cells to VS-4718 and induced apoptotic cell death upon VS-4718 treatment. Together, our results suggest that E-cadherin serves as a predictive biomarker for molecular target therapy with FAK inhibitors for patients with mesothelioma and that its expression endows MM cells with resistance to FAK inhibitors.
Collapse
|
47
|
Focal adhesion kinase depletion reduces human hepatocellular carcinoma growth by repressing enhancer of zeste homolog 2. Cell Death Differ 2017; 24:889-902. [PMID: 28338656 PMCID: PMC5423113 DOI: 10.1038/cdd.2017.34] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 02/17/2017] [Accepted: 02/23/2017] [Indexed: 12/29/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of liver cancer in humans. The focal adhesion tyrosine kinase (FAK) is often over-expressed in human HCC and FAK inhibition may reduce HCC cell invasiveness. However, the anti-oncogenic effect of FAK knockdown in HCC cells remains to be clarified. We found that FAK depletion in HCC cells reduced in vitro and in vivo tumorigenicity, by inducing G2/M arrest and apoptosis, decreasing anchorage-independent growth, and modulating the expression of several cancer-related genes. Among these genes, we showed that FAK silencing decreased transcription and nuclear localization of enhancer of zeste homolog 2 (EZH2) and its tri-methylation activity on lysine 27 of histone H3 (H3K27me3). Accordingly, FAK, EZH2 and H3K27me3 were concomitantly upregulated in human HCCs compared to non-tumor livers. In vitro experiments demonstrated that FAK affected EZH2 expression and function by modulating, at least in part, p53 and E2F2/3 transcriptional activity. Moreover, FAK silencing downregulated both EZH2 binding and histone H3K27me3 levels at the promoter of its target gene NOTCH2. Finally, we found that pharmacological inhibition of FAK activity resembled these effects although milder. In summary, we demonstrate that FAK depletion reduces HCC cell growth by affecting cancer-promoting genes including the pro-oncogene EZH2. Furthermore, we unveil a novel unprecedented FAK/EZH2 crosstalk in HCC cells, thus identifying a targetable network paving the way for new anticancer therapies.
Collapse
|
48
|
Donejko M, Rysiak E, Galicka E, Terlikowski R, Głażewska EK, Przylipiak A. Protective influence of hyaluronic acid on focal adhesion kinase activity in human skin fibroblasts exposed to ethanol. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:669-676. [PMID: 28293103 PMCID: PMC5345991 DOI: 10.2147/dddt.s125843] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Aim The aim of this study was to evaluate the effect of ethanol and hyaluronic acid (HA) on cell survival and apoptosis in cultured human skin fibroblasts. Regarding the mechanism of ethanol action on human skin fibroblasts, we investigated cell viability and apoptosis, expression of focal adhesion kinase (FAK), and the influence of HA on those processes. Materials and methods Studies were conducted in confluent human skin fibroblast cultures that were treated with 25 mM, 50 mM, and 100 mM ethanol or with ethanol and 500 µg/mL HA. Cell viability was examined using methyl thiazolyl tetrazolium (MTT) assay and NC-300 Nucleo-Counter. Imaging of the cells using a fluorescence microscope Pathway 855 was performed to measure FAK expression. Results Depending on the dosage, ethanol decreased cell viability and activated the process of apoptosis in human skin fibroblasts. HA prevented the negative influence of ethanol on cell viability and prevented apoptosis. The analysis of fluorescence imaging using BD Pathway 855 High-Content Bioimager showed the inhibition of FAK migration to the cell nucleus, depending on the increasing concentration of ethanol. Conclusion This study proves that downregulation of signaling pathway of FAK is involved in ethanol-induced apoptosis in human skin fibroblasts. The work also indicates a protective influence of HA on FAK activity in human skin fibroblasts exposed to ethanol.
Collapse
Affiliation(s)
| | - Edyta Rysiak
- Department of Medicinal Chemistry, Faculty of Pharmacy
| | | | - Robert Terlikowski
- Department of Health Restoration, Medical University of Białystok, Białystok, Poland
| | | | | |
Collapse
|
49
|
Hall JE, Schaller MD. Phospholipid binding to the FAK catalytic domain impacts function. PLoS One 2017; 12:e0172136. [PMID: 28222177 PMCID: PMC5319746 DOI: 10.1371/journal.pone.0172136] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 01/31/2017] [Indexed: 12/26/2022] Open
Abstract
Focal adhesion kinase is an essential nonreceptor tyrosine kinase that plays an important role in development, in homeostasis and in the progression of human disease. Multiple stimuli activate FAK, which requires a change in structure from an autoinhibited to activated conformation. In the autoinhibited conformation the FERM domain associates with the catalytic domain of FAK and PI(4,5)P2 binding to the FERM domain plays a role in the release of autoinhibition, activating the enzyme. An in silico model of FAK/PI(4,5)P2 interaction suggests that residues on the catalytic domain interact with PI(4,5)P2, in addition to the known FERM domain PI(4,5)P2 binding site. This study was undertaken to test the significance of this in silico observation. Mutations designed to disrupt the putative PI(4,5)P2 binding site were engineered into FAK. These mutants exhibited defects in phosphorylation and failed to completely rescue the phenotype associated with fak-/- phenotype fibroblasts demonstrating the importance of these residues in FAK function. The catalytic domain of FAK exhibited PI(4,5)P2 binding in vitro and binding activity was lost upon mutation of putative PI(4,5)P2 binding site basic residues. However, binding was not selective for PI(4,5)P2, and the catalytic domain bound to several phosphatidylinositol phosphorylation variants. The mutant exhibiting the most severe biological defect was defective for phosphatidylinositol phosphate binding, supporting the model that catalytic domain phospholipid binding is important for biochemical and biological function.
Collapse
Affiliation(s)
- Jessica E. Hall
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, West Virginia, United States of America
| | - Michael D. Schaller
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, West Virginia, United States of America
- Mary Babb Randolph Cancer Center, West Virginia University School of Medicine, Morgantown, West Virginia, United States of America
- * E-mail:
| |
Collapse
|
50
|
Panera N, Crudele A, Romito I, Gnani D, Alisi A. Focal Adhesion Kinase: Insight into Molecular Roles and Functions in Hepatocellular Carcinoma. Int J Mol Sci 2017; 18:ijms18010099. [PMID: 28067792 PMCID: PMC5297733 DOI: 10.3390/ijms18010099] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 12/23/2016] [Accepted: 12/30/2016] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death worldwide. Due to the high incidence of post-operative recurrence after current treatments, the identification of new and more effective drugs is required. In previous years, new targetable genes/pathways involved in HCC pathogenesis have been discovered through the help of high-throughput sequencing technologies. Mutations in TP53 and β-catenin genes are the most frequent aberrations in HCC. However, approaches able to reverse the effect of these mutations might be unpredictable. In fact, if the reactivation of proteins, such as p53 in tumours, holds great promise as anticancer therapy, there are studies arguing that chronic activation of these types of molecules may be deleterious. Thus, recently the efforts on potential targets have focused on actionable mutations, such as those occurring in the gene encoding for focal adhesion kinase (FAK). This tyrosine kinase, localized to cellular focal contacts, is over-expressed in a variety of human tumours, including HCC. Moreover, several lines of evidence demonstrated that FAK depletion or inhibition impair in vitro and in vivo HCC growth and metastasis. Here, we provide an overview of FAK expression and activity in the context of tumour biology, discussing the current evidence of its connection with HCC development and progression.
Collapse
Affiliation(s)
- Nadia Panera
- Liver Research Unit, Bambino Gesù Children's Hospital, IRCCS, Via S. Paolo, 15, 00146 Rome, Italy.
| | - Annalisa Crudele
- Liver Research Unit, Bambino Gesù Children's Hospital, IRCCS, Via S. Paolo, 15, 00146 Rome, Italy.
| | - Ilaria Romito
- Liver Research Unit, Bambino Gesù Children's Hospital, IRCCS, Via S. Paolo, 15, 00146 Rome, Italy.
| | - Daniela Gnani
- Liver Research Unit, Bambino Gesù Children's Hospital, IRCCS, Via S. Paolo, 15, 00146 Rome, Italy.
| | - Anna Alisi
- Liver Research Unit, Bambino Gesù Children's Hospital, IRCCS, Via S. Paolo, 15, 00146 Rome, Italy.
| |
Collapse
|