1
|
Nguyen H, Kim SH, Juang U, Gwon S, Jung W, Huang Q, Lee S, Lee B, Kwon SH, Park J. Overview of carboxyl‑terminal modulator protein 1 and its importance in various metabolic regulations (Review). Mol Med Rep 2024; 30:158. [PMID: 38994770 PMCID: PMC11258604 DOI: 10.3892/mmr.2024.13282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/21/2024] [Indexed: 07/13/2024] Open
Abstract
Acyl‑coenzyme A thioesterases (ACOTs) are crucial in mediating lipid metabolic functions, including energy expenditure, hepatic gluconeogenesis and neuronal function. The two distinct types are type I and II ACOTs, the latter of which are 'hotdog' fold superfamily members. Type II ACOTs include carboxyl‑terminal modulator protein 1 (CTMP1), also termed thioesterase superfamily member 4 (THEM4), and CTMP2, also termed THEM5. Due to their similar structural features and distinct sequence homology, CTMP1 and CTMP2 stand out from other type II ACOTs. CTMP1 was initially known as a protein kinase B (PKB) inhibitor that attenuates PKB phosphorylation. PKB is the central regulator of various cellular functions, including survival, proliferation, growth and metabolism. Therefore, by inhibiting PKB, CTMP1 can affect various cellular processes. Various other functions of CTMP1 have been revealed, including functions in cancer, brain injury, mitochondrial function and lipid metabolism. CTMP2 is a paralog of CTMP1 and was first identified as a cardiolipin remodeling factor involved in the development of fatty liver. As the functions of CTMP1 and CTMP2 were discovered separately, a review to summarize and connect these findings is essential. The current review delineates the intricate complexity of CTMP regulation across different metabolic pathways and encapsulates the principal discoveries concerning CTMP until the present day.
Collapse
Affiliation(s)
- Huonggiang Nguyen
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Seon-Hwan Kim
- Department of Neurosurgery, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Uijin Juang
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Suhwan Gwon
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Woohyeong Jung
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Qingzhi Huang
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Soohyeon Lee
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Beomwoo Lee
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - So Hee Kwon
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 21983, Republic of Korea
| | - Jongsun Park
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| |
Collapse
|
2
|
Cai A, Chen Y, Wang LS, Cusick JK, Shi Y. Depicting Biomarkers for HER2-Inhibitor Resistance: Implication for Therapy in HER2-Positive Breast Cancer. Cancers (Basel) 2024; 16:2635. [PMID: 39123362 PMCID: PMC11311605 DOI: 10.3390/cancers16152635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
HER2 (human epidermal growth factor receptor 2) is highly expressed in a variety of cancers, including breast, lung, gastric, and pancreatic cancers. Its amplification is linked to poor clinical outcomes. At the genetic level, HER2 is encoded by the ERBB2 gene (v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 2), which is frequently mutated or amplified in cancers, thus spurring extensive research into HER2 modulation and inhibition as viable anti-cancer strategies. An impressive body of FDA-approved drugs, including anti-HER2 monoclonal antibodies (mAbs), antibody-drug conjugates (ADCs), and HER2-tyrosine kinase inhibitors (TKIs), have demonstrated success in enhancing overall survival (OS) and disease progression-free survival (PFS). Yet, drug resistance remains a persistent challenge and raises the risks of metastatic potential and tumor relapse. Research into alternative therapeutic options for HER2+ breast cancer therefore proves critical for adapting to this ever-evolving landscape. This review highlights current HER2-targeted therapies, discusses predictive biomarkers for drug resistance, and introduces promising emergent therapies-especially combination therapies-that are aimed at overcoming drug resistance in the context of HER2+ breast cancer.
Collapse
Affiliation(s)
- Alvan Cai
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (A.C.); (J.K.C.)
| | - Yuan Chen
- Section Pathology of the Institute of Forensic Medicine, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany;
| | - Lily S. Wang
- University of California, Berkeley, CA 94720, USA;
| | - John K. Cusick
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (A.C.); (J.K.C.)
| | - Yihui Shi
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (A.C.); (J.K.C.)
- California Pacific Medical Center Research Institute, Sutter Bay Hospitals, San Francisco, CA 94107, USA
| |
Collapse
|
3
|
Xie W, Liu W, Wang L, Zhu B, Zhao C, Liao Z, Li Y, Jiang X, Liu J, Ren C. Roles of THEM4 in the Akt pathway: a double-edged sword. J Zhejiang Univ Sci B 2024; 25:541-556. [PMID: 39011675 PMCID: PMC11254685 DOI: 10.1631/jzus.b2300457] [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: 06/26/2023] [Accepted: 10/10/2023] [Indexed: 07/17/2024]
Abstract
The protein kinase B (Akt) pathway can regulate the growth, proliferation, and metabolism of tumor cells and stem cells through the activation of multiple downstream target genes, thus affecting the development and treatment of a range of diseases. Thioesterase superfamily member 4 (THEM4), a member of the thioesterase superfamily, is one of the Akt kinase-binding proteins. Some studies on the mechanism of cancers and other diseases have shown that THEM4 binds to Akt to regulate its phosphorylation. Initially, THEM4 was considered an endogenous inhibitor of Akt, which can inhibit the phosphorylation of Akt in diseases such as lung cancer, pancreatic cancer, and liver cancer, but subsequently, THEM4 was shown to promote the proliferation of tumor cells by positively regulating Akt activity in breast cancer and nasopharyngeal carcinoma, which contradicts previous findings. Considering these two distinct views, this review summarizes the important roles of THEM4 in the Akt pathway, focusing on THEM4 as an Akt-binding protein and its regulatory relationship with Akt phosphorylation in various diseases, especially cancer. This work provides a better understanding of the roles of THEM4 combined with Akt in the treatment of diseases.
Collapse
Affiliation(s)
- Wen Xie
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Weidong Liu
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Lei Wang
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Bin Zhu
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Cong Zhao
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Ziling Liao
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Yihan Li
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Xingjun Jiang
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jie Liu
- Department of Critical Care Medicine, Hainan Hospital of Chinese PLA General Hospital, Hainan Province Clinical Medical Center, Sanya 572013, China. ,
| | - Caiping Ren
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China.
| |
Collapse
|
4
|
Bai L, Yang P, Han B, Kong L. Progress of the acyl-Coenzyme A thioester hydrolase family in cancer. Front Oncol 2024; 14:1374094. [PMID: 38562172 PMCID: PMC10982514 DOI: 10.3389/fonc.2024.1374094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
In recent years, the acyl-Coenzyme A thioester hydrolase family (ACOTs) has received wide attention as a key link in lipid metabolism. This family is a class of enzymes that catalyze the hydrolysis of fatty acyl-Coenzyme A, disrupting the thioester bond present within acyl-CoA ester molecules to produce free fatty acids (FFA) and the corresponding coenzyme A (CoA). Such enzymes play a very important role in lipid metabolism through maintaining appropriate levels of intracellular FFA and fatty acyl-CoA as well as CoA. It is broadly divided into two distinct subgroups, the type-I α/β-hydrolase fold enzyme superfamily and the type-II 'hot dog' fold superfamily. There are currently four human type-I genes and eight human type-II genes. Although the two subgroups catalyze the same reaction, they are not structurally similar, do not share the same sequence homology, and differ greatly in protein executive functions. This review summarizes the classification of the acyl-CoA thioester hydrolase family, an overview of the structural sequences, and advances in digestive, respiratory, and urinary systemic tumors. In order to explore potential specific drug targets and effective interventions, to provide new strategies for tumor prevention and treatment.
Collapse
Affiliation(s)
- Lu Bai
- Inner Mongolia Medical University, Hohhot, China
- Department of Pathology, Peking University Cancer Hospital & Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Pengjie Yang
- Thoracic Surgery Department, Peking University Cancer Hospital & Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Bater Han
- Thoracic Surgery Department, Peking University Cancer Hospital & Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Linghui Kong
- Department of Pathology, Peking University Cancer Hospital & Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
5
|
El-Khazragy N, Gaballah A, Bakkar A, Hemida EHA, Samir N, Tarek M, Adly HM, Saleh SAK, Hanna DH. PTEN rs701848 Polymorphism is Associated with Trastuzumab Resistance in HER2-positive Metastatic Breast Cancer and Predicts Progression-free Survival. Clin Breast Cancer 2023; 23:e131-e139. [PMID: 36599770 DOI: 10.1016/j.clbc.2022.12.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/06/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Trastuzumab is an effective therapeutic approach for HER2-positive metastatic breast cancer (BC). However, a considerable number of patients develop resistance along the course of the disease. PTEN rs701848 polymorphisms are associated with an increased risk of developing cancer and have a potential role in predicting drug resistance. OBJECTIVE We studied the significance of PTEN rs701848 variants as significant predictors for trastuzumab resistance in HER2-positive metastatic BC patients. Therefore, considering their value in predicting clinical outcomes. MATERIALS AND METHODS This case-control study was conducted among female patients with HER2-positive metastatic breast cancer who underwent Trastuzumab therapy during the period from March 2017 to December 2020. PTEN rs701848 genotypes were analyzed in 160 HER2-positive metastatic breast cancer who received Trastuzumab therapy and clinically monitored for therapeutic response. RESULTS PTEN rs701848 is deemed a significant predictor of Trastuzumab resistance and an independent prognostic factor of progression-free survival (PPFS). In particular, the C allele is associated with increased risk for Trastuzumab resistance and shorter PFS as compared to the homozygous TT genotype. CONCLUSION PTEN rs701848 is significant predictor of trastuzumab resistance. Therefore, their value in predicting clinical outcomes is recommended.
Collapse
Affiliation(s)
- Nashwa El-Khazragy
- Department of Clinical Pathology-Hematology and Ain Shams Medical Research Institute (MASRI), Faculty of Medicine, Ain Shams University, Cairo, Egypt; Department of Genetics and Molecular Biology, Egypt Center for Research and Regenerative Medicine (ECRRM), Cairo, Egypt.
| | - Ahmed Gaballah
- Department of Clinical Oncology and Nuclear Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ashraf Bakkar
- Faculty of Biotechnology, October University for Modern Sciences and Arts, Giza, Egypt
| | - Eman H A Hemida
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Nehal Samir
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Marwa Tarek
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Heba M Adly
- Department of Community Medicine and Pilgrims Healthcare, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Saleh A K Saleh
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Demiana H Hanna
- Department of Biochemistry, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
6
|
Lin CH, Lin WD, Huang YC, Chen YC, Loh ZJ, Ger LP, Lin FC, Li HY, Cheng HC, Lee KH, Hsiao M, Lu PJ. Carboxyl-terminal modulator protein facilitates tumor metastasis in triple-negative breast cancer. Cancer Gene Ther 2023; 30:404-413. [PMID: 36400965 PMCID: PMC10014580 DOI: 10.1038/s41417-022-00559-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/18/2022] [Accepted: 10/28/2022] [Indexed: 11/19/2022]
Abstract
Currently, the survival rate for breast cancer is more than 90%, but once the cancer cells metastasize to distal organs, the survival rate is dramatically reduced, to less than 30%. Triple-negative breast cancer accounts for 15-20% of all breast cancers. Triple-negative breast cancer (TNBC) is associated with poor prognostic and diagnostic outcomes due to the limiting therapeutic strategies, relative to non-TNBC breast cancers. Therefore, the development of targeted therapy for TNBC metastasis remains an urgent issue. In this study, high Carboxyl-terminal modulator protein (CTMP) is significantly associated with recurrence and disease-free survival rate in TNBC patients. Overexpression of CTMP promotes migration and invasion abilities in BT549 cells. Down-regulating of CTMP expression inhibits migration and invasion abilities in MDA-MB-231 cells. In vivo inoculation of high-CTMP cells enhances distant metastasis in mice. The metastasis incidence rate is decreased in mice injected with CTMP-downregulating MDA-MB-231 cells. Gene expression microarray analysis indicates the Akt-dependent pathway is significantly enhanced in CTMP overexpressing cells compared to the parental cells. Blocking Akt activation via Akt inhibitor treatment or co-expression of the dominant-negative form of Akt proteins successfully abolishes the CTMP mediating invasion in TNBC cells. Our findings suggest that CTMP is a potential diagnostic marker for recurrence and poor disease-free survival in TNBC patients. CTMP promotes TNBC metastasis via the Akt-activation-dependent pathway.
Collapse
Affiliation(s)
- Cheng-Han Lin
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan
| | - Wen-Der Lin
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan
| | - Yun-Chin Huang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan
| | - Yu-Chia Chen
- Division of General Surgery, Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung, 813414, Taiwan
| | - Zhu-Jun Loh
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan
| | - Luo-Ping Ger
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, 813414, Taiwan
| | - Forn-Chia Lin
- Department of Radiation Oncology, National Cheng Kung University Hospital, Tainan, 70401, Taiwan
| | - Hao-Yi Li
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan
| | - Hui-Chuan Cheng
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan
| | - Kuen-Haur Lee
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Pei-Jung Lu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan. .,Department of Clinical Medicine Research, National Cheng Kung University Hospital, Tainan, 70401, Taiwan.
| |
Collapse
|
7
|
Chantada-Vázquez MDP, Conde-Amboage M, Graña-López L, Vázquez-Estévez S, Bravo SB, Núñez C. Circulating Proteins Associated with Response and Resistance to Neoadjuvant Chemotherapy in HER2-Positive Breast Cancer. Cancers (Basel) 2022; 14:cancers14041087. [PMID: 35205837 PMCID: PMC8870308 DOI: 10.3390/cancers14041087] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/24/2022] [Accepted: 02/16/2022] [Indexed: 12/13/2022] Open
Abstract
Simple Summary The goal of this study was to find circulating proteins that can be easily sampled and incorporated into a clinical setting to improve predictive treatment response in HER2-positive breast cancer patients receiving neoadjuvant chemotherapy. We looked for potential biomarkers in serum, which we identified using two proteomics techniques: qualitative LC-MS/MS and a quantitative assay that assessed protein expression between responders and non-responders HER2-positive breast cancer patients to neoadjuvant chemotherapy. Abstract Despite the increasing use of neoadjuvant chemotherapy (NAC) in HER2-positive breast cancer (BC) patients, the clinical problem of predicting individual treatment response remains unanswered. Furthermore, the use of ineffective chemotherapeutic regimens should be avoided. Serum biomarker levels are being studied more and more for their ability to predict therapy response and aid in the development of personalized treatment regimens. This study aims to identify effective protein networks and biomarkers to predict response to NAC in HER2-positive BC patients through an exhaustive large-scale LC-MS/MS-based qualitative and quantitative proteomic profiling of serum samples from responders and non-responders. Serum samples from HER2-positive BC patients were collected before NAC and were processed by three methods (with and without nanoparticles). The qualitative analysis revealed differences in the proteomic profiles between responders and non-responders, mainly in proteins implicated in the complement and coagulation cascades and apolipoproteins. Qualitative analysis confirmed that three proteins (AFM, SERPINA1, APOD) were correlated with NAC resistance. In this study, we show that serum biomarker profiles can predict treatment response and outcome in the neoadjuvant setting. If these findings are further developed, they will be of significant clinical utility in the design of treatment regimens for individual BC patients.
Collapse
Affiliation(s)
- María del Pilar Chantada-Vázquez
- Research Unit, Lucus Augusti University Hospital (HULA), Servizo Galego de Saúde (SERGAS), 27002 Lugo, Spain;
- Proteomic Unit, Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago de Compostela (CHUS), 15706 Santiago de Compostela, Spain
| | - Mercedes Conde-Amboage
- Models of Optimization Decision, Statistics and Applications Research Group (MODESTYA), Department of Statistics, Mathematical Analysis and Optimization, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain;
- CITMAga, 15782 Santiago de Compostela, Spain
| | - Lucía Graña-López
- Breast Pathology Group, Lucus Augusti University Hospital (HULA)-IDIS, Servizo Galego de Saúde (SERGAS), 27002 Lugo, Spain;
- Radiology Department, Lucus Augusti University Hospital (HULA), Servizo Galego de Saúde (SERGAS), 27002 Lugo, Spain
| | - Sergio Vázquez-Estévez
- Oncology Division, Lucus Augusti University Hospital (HULA), Servizo Galego de Saúde (SERGAS), 27002 Lugo, Spain;
| | - Susana B. Bravo
- Proteomic Unit, Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago de Compostela (CHUS), 15706 Santiago de Compostela, Spain
- Correspondence: (S.B.B.); (C.N.)
| | - Cristina Núñez
- Research Unit, Lucus Augusti University Hospital (HULA), Servizo Galego de Saúde (SERGAS), 27002 Lugo, Spain;
- Correspondence: (S.B.B.); (C.N.)
| |
Collapse
|
8
|
Kaboli PJ, Imani S, Jomhori M, Ling KH. Chemoresistance in breast cancer: PI3K/Akt pathway inhibitors vs the current chemotherapy. Am J Cancer Res 2021; 11:5155-5183. [PMID: 34765318 PMCID: PMC8569340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 06/12/2021] [Indexed: 06/13/2023] Open
Abstract
Breast cancer is the most prevalent type of cancer among women. Several types of drugs, targeting the specific proteins expressed on the breast cancer cell surface (such as receptor tyrosine kinases and immune checkpoint regulators) and proteins involved in cell cycle and motility (including cyclin-dependent kinases, DNA stabilisers, and cytoskeleton modulators) are approved for different subtypes of breast cancer. However, breast cancer also has a poor response to conventional chemotherapy due to intrinsic and acquired resistance, and an Akt fingerprint is detectable in most drug-resistant cases. Overactivation of Akt and its upstream and downstream regulators in resistant breast cancer cells is considered a major potential target for novel anti-cancer therapies, suggesting that Akt signalling acts as a cellular mechanism against chemotherapy. The present review has shown that sustained activation of Akt results in resistance to different types of chemotherapy. Akt signalling plays a cellular defence role against chemotherapy and (1) enhances multi-drug resistance, (2) increases reactive oxygen species at breast tumor microenvironment, (3) enhances anaerobic metabolism, (4) inhibits the tricarboxylic cycle, (5) promotes PD-L1 upregulation, (6) inhibits apoptosis, (7) increases glucose uptake, and more importantly (8) recruits and interconnects the plasma membrane, nucleus, endoplasmic reticulum, and mitochondria to hijack breast cancer cells and rescue these cells from chemotherapy. Therefore, Akt signalling is considered a cellular defence mechanism employed against chemotherapeutic effects. In addition, interfering roles of PI3K/Akt signalling on the current cytotoxic and molecularly targeted therapy as well as immunotherapy of breast cancer are discussed with a clinical approach. Although, alpelisib, a PIK3CA inhibitor, is the only PI3K/Akt pathway inhibitor approved for breast cancer, we also highlight well-evaluated inhibitors of PI3K/Akt signalling based on different subtypes of breast cancer, which are under clinical trials whether as monotherapy or in combination with other types of chemotherapy.
Collapse
Affiliation(s)
- Parham Jabbarzadeh Kaboli
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology, and Center for Molecular Medicine, China Medical UniversityTaichung 404, Taiwan
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra MalaysiaSerdang, Selangor 43400, Malaysia
| | - Saber Imani
- Department of Oncology, The Affiliated Hospital of Southwest Medical UniversityLuzhou, Sichuan 646000, P. R. China
| | - Masume Jomhori
- Department of Biotechnology Research, Razi Vaccine and Serum Research InstituteMashhad, Iran
| | - King-Hwa Ling
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra MalaysiaSerdang, Selangor 43400, Malaysia
- Department of Genetics, Harvard Medical SchoolBoston, MA 02115, USA
| |
Collapse
|
9
|
Zhang S, Li D, Zhao M, Yang F, Sang C, Yan C, Wang Z, Li Y. Exosomal miR-183-5p Shuttled by M2 Polarized Tumor-Associated Macrophage Promotes the Development of Colon Cancer via Targeting THEM4 Mediated PI3K/AKT and NF-κB Pathways. Front Oncol 2021; 11:672684. [PMID: 34249713 PMCID: PMC8267908 DOI: 10.3389/fonc.2021.672684] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/10/2021] [Indexed: 12/24/2022] Open
Abstract
Background Abnormal accumulation of macrophages in the colon cancer (CC) contribute to its progression. miR-183-5p has been confirmed as an oncogene in CC and this article explores the effect and mechanism of exosomal miR-183-5p enriched by M2-polarized tumor-associated macrophages (TAM) on CC cells. Methods The human macrophage THP1 was induced to M2 polarization through IL-4 and IL-13 treatment. Exosomes in THP1 were isolated through ultracentrifugation, and the miR-183-5p expression in macrophages and exosomes was verified by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). The miR-183-5p inhibitors and mimics were applied to down-regulate and upregulate miR-183-5p in macrophages, respectively. Meanwhile, CC cell lines LoVo and SW480 were treated with the macrophage conditioned medium and exosomes, respectively. CC cells’ proliferation, invasion, and apoptosis were tested by the cell counting kit-8 (CCK-8) assay, colony formation assay, flow cytometry (FCM), Transwell assay, and xenograft assay, respectively. The profiles of thioesterase superfamily member 4 (THEM4), Akt, and NF-κB were compared by Western blotting (WB). Results The miR-183-5p level in M2-TAM and M2-TAM-derived exosomes was significantly increased. Meanwhile, M2-TAM and M2-TAM-derived exosomes significantly facilitated CC cell proliferation and invasion and dampened apoptosis. Overexpression of miR-183-5p in M2-TAM aggravated M2-TAM-mediated promotive effects on CC cells, with down-regulating miR-183-5p reversed M2-TAM-mediated tumor-promotive effects. Mechanically, miR-183-5p targeted THEM4 and inhibited its mRNA and protein expression. Overexpressing THEM4 abated miR-183-5p-mediated carcinogenic effects and inactivates Akt and NF-κB pathways in CC cells. Overall, this article elaborated that exosomal miR-183-5p shuttled by M2-TAM mediated Akt/NF-κB pathway to accelerate CC progression through targeting THEM4.
Collapse
Affiliation(s)
- Shangxin Zhang
- Department of Gastrointestinal Surgery & Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Deguan Li
- Department of Gastrointestinal Surgery & Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Min Zhao
- Department of General Surgery, Yanqing District Hospital (Peking University Third Hospital Yanqing Hospital), Beijing, China
| | - Fei Yang
- Department of Orthopedics, Beijing Yanqing District Hospital (Peking University Third Hospital Yanqing Hospital), Beijing, China
| | - Changye Sang
- Department of General Surgery, Yanqing District Hospital (Peking University Third Hospital Yanqing Hospital), Beijing, China
| | - Changhong Yan
- Department of General Surgery, Yanqing District Hospital (Peking University Third Hospital Yanqing Hospital), Beijing, China
| | - Zhenjun Wang
- Department of General Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yongxiang Li
- Department of Gastrointestinal Surgery & Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
10
|
Jafarzadeh L, Khakpoor-Koosheh M, Mirzaei H, Mirzaei HR. Biomarkers for predicting the outcome of various cancer immunotherapies. Crit Rev Oncol Hematol 2021; 157:103161. [DOI: 10.1016/j.critrevonc.2020.103161] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 10/21/2020] [Accepted: 11/05/2020] [Indexed: 12/11/2022] Open
|
11
|
Silva-Oliveira R, Pereira FF, Petronilho S, Martins AT, Lameirinhas A, Constâncio V, Caldas-Ribeiro I, Salta S, Lopes P, Antunes L, Castro F, de Sousa SP, Henrique R, Jerónimo C. Clinical Significance of ARID1A and ANXA1 in HER-2 Positive Breast Cancer. J Clin Med 2020; 9:E3911. [PMID: 33276477 PMCID: PMC7761245 DOI: 10.3390/jcm9123911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/21/2020] [Accepted: 12/01/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND trastuzumab is considered the standard of care for human epidermal growth factor receptor-2 (HER-2+) breast cancer patients. Regardless of the benefits of its use, many early-stage patients eventually recur, and usually, the disease progresses within a year. Since about half of the HER-2+ patients do not respond to trastuzumab, new biomarkers of prognosis and prediction are warranted to allow a better patient stratification. Annexin A1 (ANXA1) was previously reported to contribute to trastuzumab resistance through AKT activation. An association between adenine thymine-rich interactive domain 1A (ARID1A) loss and ANXA1 upregulation was also previously suggested by others. METHODS in this study, we examined tissue samples from 215 HER-2+ breast cancer patients to investigate the value of ARID1A and ANXA1 protein levels in trastuzumab response prediction and patient outcome. Expression of ARID1A and ANXA1 were assessed by immunohistochemistry. RESULTS contrary to what was expected, no inverse association was found between ARID1A and ANXA1 expression. HER-2+ (non-luminal) tumours displayed higher ANXA1 expression than luminal B-like (HER-2+) tumours. Concerning trastuzumab resistance, ARID1A and ANXA1 proteins did not demonstrate predictive value as biomarkers. Nevertheless, an association was depicted between ANXA1 expression and breast cancer mortality and relapse. CONCLUSIONS overall, our results suggest that ANXA1 may be a useful prognostic marker in HER-2+ patients. Additionally, its ability to discriminate between HER-2+ (non-luminal) and luminal B-like (HER-2+) patients might assist in patient stratification regarding treatment strategy.
Collapse
Affiliation(s)
- Rita Silva-Oliveira
- Cancer Biology & Epigenetics Group—Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (R.S.-O.); (S.P.); (A.T.M.); (A.L.); (V.C.); (I.C.-R.); (S.S.); (P.L.); (R.H.)
- Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua de Jorge Viterbo Ferreira n. 228, 4050-313 Porto, Portugal
| | - Filipa Ferreira Pereira
- Breast Cancer Clinic and Department of Medical Oncology, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (F.F.P.); (S.P.d.S.)
| | - Sara Petronilho
- Cancer Biology & Epigenetics Group—Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (R.S.-O.); (S.P.); (A.T.M.); (A.L.); (V.C.); (I.C.-R.); (S.S.); (P.L.); (R.H.)
- Department of Pathology, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Ana Teresa Martins
- Cancer Biology & Epigenetics Group—Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (R.S.-O.); (S.P.); (A.T.M.); (A.L.); (V.C.); (I.C.-R.); (S.S.); (P.L.); (R.H.)
- Department of Pathology, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Ana Lameirinhas
- Cancer Biology & Epigenetics Group—Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (R.S.-O.); (S.P.); (A.T.M.); (A.L.); (V.C.); (I.C.-R.); (S.S.); (P.L.); (R.H.)
| | - Vera Constâncio
- Cancer Biology & Epigenetics Group—Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (R.S.-O.); (S.P.); (A.T.M.); (A.L.); (V.C.); (I.C.-R.); (S.S.); (P.L.); (R.H.)
| | - Inês Caldas-Ribeiro
- Cancer Biology & Epigenetics Group—Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (R.S.-O.); (S.P.); (A.T.M.); (A.L.); (V.C.); (I.C.-R.); (S.S.); (P.L.); (R.H.)
| | - Sofia Salta
- Cancer Biology & Epigenetics Group—Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (R.S.-O.); (S.P.); (A.T.M.); (A.L.); (V.C.); (I.C.-R.); (S.S.); (P.L.); (R.H.)
| | - Paula Lopes
- Cancer Biology & Epigenetics Group—Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (R.S.-O.); (S.P.); (A.T.M.); (A.L.); (V.C.); (I.C.-R.); (S.S.); (P.L.); (R.H.)
- Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua de Jorge Viterbo Ferreira n. 228, 4050-313 Porto, Portugal
| | - Luís Antunes
- Cancer Epidemiology Group—Research Center & Department of Epidemiology, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal;
| | - Fernando Castro
- Breast Cancer Clinic and Department of Surgical Oncology, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal;
| | - Susana Palma de Sousa
- Breast Cancer Clinic and Department of Medical Oncology, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (F.F.P.); (S.P.d.S.)
| | - Rui Henrique
- Cancer Biology & Epigenetics Group—Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (R.S.-O.); (S.P.); (A.T.M.); (A.L.); (V.C.); (I.C.-R.); (S.S.); (P.L.); (R.H.)
- Department of Pathology, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua de Jorge Viterbo Ferreira n. 228, 4050-313 Porto, Portugal
| | - Carmen Jerónimo
- Cancer Biology & Epigenetics Group—Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (R.S.-O.); (S.P.); (A.T.M.); (A.L.); (V.C.); (I.C.-R.); (S.S.); (P.L.); (R.H.)
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua de Jorge Viterbo Ferreira n. 228, 4050-313 Porto, Portugal
| |
Collapse
|
12
|
Mocenigo M, Porchetta A, Rossetti M, Brass E, Tonini L, Puzzi L, Tagliabue E, Triulzi T, Marini B, Ricci F, Ippodrino R. Rapid, Cost-Effective Peptide/Nucleic Acid-Based Platform for Therapeutic Antibody Monitoring in Clinical Samples. ACS Sens 2020; 5:3109-3115. [PMID: 32909731 DOI: 10.1021/acssensors.0c01046] [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] [Indexed: 02/07/2023]
Abstract
We demonstrate here a homogeneous assay, named NanoHybrid, for monoclonal antibody quantification directly in serum samples in a single-step format. NanoHybrid is composed of both synthetic peptide nucleic acids (PNAs) and nucleic acid strands conjugated to recognition elements and optical labels and is designed to allow fast fluorescence quantification of a therapeutic antibody. More specifically, we have characterized our analytical assay for the detection of trastuzumab (Herceptin), a monoclonal antibody (mAb) drug used for breast cancer treatment and for tumors overexpressing the HER2/neu protein. We show here that NanoHybrid is capable of performing fast drug quantification directly in blood serum. The results obtained with a pool of samples from breast cancer patients under trastuzumab treatment are compared with CE-IVD ELISA (enzyme-linked immunosorbent assay) showing a good agreement (Cohen's K = 0.729). Due to the modular nature of the NanoHybrid platform, this technology can be programmed to potentially detect and quantify any antibody for which a high-affinity recognition element has been characterized. We envision the application of NanoHybrid in a point-of-care (POC) drug monitoring system based on disposable kits for therapeutic drug management.
Collapse
Affiliation(s)
- Marco Mocenigo
- Ulisse BioMed Labs, Area Science Park, SS 14, km 163.5, 34149 Trieste, Italy
- Molecular Genetics and Biotechnology PhD Study Programme, University of Nova Gorica, Vipavska 13, 5000 Nova Gorica, Slovenia
| | - Alessandro Porchetta
- Department of Chemistry, University of Rome, Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Marianna Rossetti
- Department of Chemistry, University of Rome, Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Erik Brass
- Ulisse BioMed Labs, Area Science Park, SS 14, km 163.5, 34149 Trieste, Italy
| | - Lucia Tonini
- Ulisse BioMed Labs, Area Science Park, SS 14, km 163.5, 34149 Trieste, Italy
| | - Luca Puzzi
- Ulisse BioMed Labs, Area Science Park, SS 14, km 163.5, 34149 Trieste, Italy
| | - Elda Tagliabue
- Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Tiziana Triulzi
- Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Bruna Marini
- Ulisse BioMed Labs, Area Science Park, SS 14, km 163.5, 34149 Trieste, Italy
| | - Francesco Ricci
- Department of Chemistry, University of Rome, Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Rudy Ippodrino
- Ulisse BioMed Labs, Area Science Park, SS 14, km 163.5, 34149 Trieste, Italy
| |
Collapse
|
13
|
Wang B, Xu X, Liu X, Wang D, Zhuang H, He X, Han T, Hong J. Enolase-phosphatase 1 acts as an oncogenic driver in glioma. J Cell Physiol 2020; 236:1184-1194. [PMID: 32654229 DOI: 10.1002/jcp.29926] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 07/01/2020] [Indexed: 12/15/2022]
Abstract
Enolase-phosphatase 1 (ENOPH1), a newly identified enzyme involved in l-methionine biosynthesis, is associated with anxiety and depression. In this study, ENOPH1 was found to play a crucial role in promoting the proliferation and migration of glioma cells. Among high-grade glioma patients, the overall survival of the group showing high ENOPH1 expression was shorter than that of the group showing low ENOPH1 expression. ENOPH1 knockdown inhibited glioma cell proliferation and migration. In parallel, ENOPH1 knockdown suppressed tumor growth capacity and prolonged survival in an orthotopic glioma model. Mechanistically, we found that ENOPH1 activates the PI3K/AKT/mTOR signaling pathway by regulating THEM4. In conclusion, ENOPH1 is an important mediator that promotes glioma cell proliferation and migration.
Collapse
Affiliation(s)
- Bo Wang
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative diseases, Tianjin Neurosurgical Institute, Tianjin, China.,State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Xin Xu
- Department of Neurosurgery, Xuanwu Hospital, International Neuroscience Institute, Capital Medical University, Beijing, China
| | - Xi Liu
- Department of Gastroenterology, Tianjin Nankai Hospital, Tianjin, China
| | - Dong Wang
- Department of Neurosurgery, Tianjin Medical University, General Hospital, Tianjin Key Laboratory of Injuries, Variations, and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
| | - Hao Zhuang
- Department of Hepatic Biliary Pancreatic Surgery, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Xin He
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia.,Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Tong Han
- Department of Medical Imaging, Tianjin Huanhu Hospital, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative diseases, Tianjin Neurosurgical Institute, Tianjin, China
| | - Jian Hong
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative diseases, Tianjin Neurosurgical Institute, Tianjin, China
| |
Collapse
|
14
|
Urbano N, Scimeca M, Bonfiglio R, Bonanno E, Schillaci O. New advance in breast cancer pathology and imaging. Future Oncol 2019; 15:2707-2722. [DOI: 10.2217/fon-2019-0017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The improvement of knowledge concerning the pathology of breast cancer could provide the rationale for the development of new imaging diagnostic protocols. Indeed, as for the microcalcifications, new histopathological markers can be used as target for in vivo early detection of breast cancer lesions by using molecular imaging techniques such as positron emission tomography. Specifically, the mutual contribution of these medical specialties can ‘nourish’ the dream of a personalized medicine that takes into account the intrinsic variability of breast cancer. In this review, we report the main discoveries concerning breast cancer pathology highlighting the possible cooperation between the departments of anatomic pathology and imaging diagnostics.
Collapse
Affiliation(s)
- Nicoletta Urbano
- Nuclear Medicine, Policlinico ‘Tor Vergata,’ viale Oxford, 81, Rome, 00133, Italy
| | - Manuel Scimeca
- Department of Biomedicine & Prevention, University of Rome ‘Tor Vergata’, Via Montpellier 1, Rome 00133, Italy
- IRCCS San Raffaele, Via di Val Cannuta 247, 00166, Rome, Italy
- Fondazione Umberto Veronesi (FUV), Piazza Velasca 5, 20122 Milano (Mi), Italy
| | - Rita Bonfiglio
- Department of Experimental Medicine, University ‘Tor Vergata’, Via Montpellier 1, Rome 00133, Italy
| | - Elena Bonanno
- Department of Experimental Medicine, University ‘Tor Vergata’, Via Montpellier 1, Rome 00133, Italy
- Neuromed Group, ‘Diagnostica Medica’ & ‘Villa dei Platani', Via Errico Carmelo, 2, 83100 Avellino AV, Italy
| | - Orazio Schillaci
- Department of Biomedicine & Prevention, University of Rome ‘Tor Vergata’, Via Montpellier 1, Rome 00133, Italy
- IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
15
|
Scimeca M, Urbano N, Bonfiglio R, Duggento A, Toschi N, Schillaci O, Bonanno E. Novel insights into breast cancer progression and metastasis: A multidisciplinary opportunity to transition from biology to clinical oncology. Biochim Biophys Acta Rev Cancer 2019; 1872:138-148. [PMID: 31348975 DOI: 10.1016/j.bbcan.2019.07.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/25/2019] [Accepted: 07/15/2019] [Indexed: 12/11/2022]
Abstract
According to the most recent epidemiological studies, breast cancer shows the highest incidence and the second leading cause of death in women. Cancer progression and metastasis are the main events related to poor survival of breast cancer patients. This can be explained by the presence of highly resistant to chemo- and radiotherapy stem cells in many breast tumor tissues. In this context, numerous studies highlighted the possible involvement of epithelial to mesenchymal transition phenomenon as biological program to generate cancer stem cells, and thus participate to both metastatic and drug resistance process. Therefore, the comprehension of mechanisms (both cellular and molecular) involved in breast cancer occurrence and progression can lay the foundation for the development of new diagnostic and therapeutical protocols. In this review, we reported the most important findings in the field of breast cancer highlighting the most recent data concerning breast tumor biology, diagnosis and therapy.
Collapse
Affiliation(s)
- Manuel Scimeca
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Via Montpellier 1, Rome 00133, Italy; San Raffaele University, Via di Val Cannuta 247, 00166 Rome, Italy; Fondazione Umberto Veronesi (FUV), Piazza Velasca 5, 20122 Milano (Mi), Italy.
| | | | - Rita Bonfiglio
- Department of Experimental Medicine, University "Tor Vergata", Via Montpellier 1, Rome 00133, Italy
| | - Andrea Duggento
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Via Montpellier 1, Rome 00133, Italy
| | - Nicola Toschi
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Via Montpellier 1, Rome 00133, Italy; Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging and Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Via Montpellier 1, Rome 00133, Italy; IRCCS Neuromed, Pozzilli, Italy
| | - Elena Bonanno
- Department of Experimental Medicine, University "Tor Vergata", Via Montpellier 1, Rome 00133, Italy; Neuromed Group, "Diagnostica Medica" and "Villa dei Platani", Avellino, Italy
| |
Collapse
|
16
|
Liu X, Yang Q, Zhu LH, Liu J, Deng KQ, Zhu XY, Liu Y, Gong J, Zhang P, Li S, Xia H, She ZG. Carboxyl-Terminal Modulator Protein Ameliorates Pathological Cardiac Hypertrophy by Suppressing the Protein Kinase B Signaling Pathway. J Am Heart Assoc 2018; 7:JAHA.118.008654. [PMID: 29945911 PMCID: PMC6064906 DOI: 10.1161/jaha.118.008654] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background Carboxyl‐terminal modulator protein (CTMP) has been implicated in cancer, brain injury, and obesity. However, the role of CTMP in pathological cardiac hypertrophy has not been identified. Methods and Results In this study, decreased expression of CTMP was observed in both human failing hearts and murine hypertrophied hearts. To further explore the potential involvement of CTMP in pathological cardiac hypertrophy, cardiac‐specific CTMP knockout and overexpression mice were generated. In vivo experiments revealed that CTMP deficiency exacerbated the cardiac hypertrophy, fibrosis, and function induced by pressure overload, whereas CTMP overexpression alleviated the response to hypertrophic stimuli. Consistent with the in vivo results, adenovirus‐mediated gain‐of‐function or loss‐of‐function experiments showed that CTMP also exerted a protective effect against hypertrophic responses to angiotensin II in vitro. Mechanistically, CTMP ameliorated pathological cardiac hypertrophy through the blockade of the protein kinase B signaling pathway. Moreover, inhibition of protein kinase B activation with LY294002 rescued the deteriorated effect in aortic banding–treated cardiac‐specific CTMP knockout mice. Conclusions Taken together, these findings imply, for the first time, that increasing the cardiac expression of CTMP may be a novel therapeutic strategy for pathological cardiac hypertrophy.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cells, Cultured
- Disease Models, Animal
- Fibrosis
- Humans
- Hypertrophy, Left Ventricular/enzymology
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/prevention & control
- Male
- Membrane Proteins/metabolism
- Mice, Knockout
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Palmitoyl-CoA Hydrolase
- Proto-Oncogene Proteins c-akt/metabolism
- Rats, Sprague-Dawley
- Signal Transduction
- Thiolester Hydrolases/metabolism
- Ventricular Function, Left
- Ventricular Remodeling
Collapse
Affiliation(s)
- Xiaoxiong Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Qin Yang
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Li-Hua Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jia Liu
- Department of Cardiology, First Hospital of Jilin University, Changchun, China
| | - Ke-Qiong Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Xue-Yong Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Ye Liu
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Jun Gong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Peng Zhang
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Shuyan Li
- Department of Cardiology, First Hospital of Jilin University, Changchun, China
| | - Hao Xia
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| |
Collapse
|
17
|
Wang Y, Guan A, Wickramasekara S, Phillips KS. Analytical Chemistry in the Regulatory Science of Medical Devices. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2018; 11:307-327. [PMID: 29579404 DOI: 10.1146/annurev-anchem-061417-125556] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In the United States, regulatory science is the science of developing new tools, standards, and approaches to assess the safety, efficacy, quality, and performance of all Food and Drug Administration-regulated products. Good regulatory science facilitates consumer access to innovative medical devices that are safe and effective throughout the Total Product Life Cycle (TPLC). Because the need to measure things is fundamental to the regulatory science of medical devices, analytical chemistry plays an important role, contributing to medical device technology in two ways: It can be an integral part of an innovative medical device (e.g., diagnostic devices), and it can be used to support medical device development throughout the TPLC. In this review, we focus on analytical chemistry as a tool for the regulatory science of medical devices. We highlight recent progress in companion diagnostics, medical devices on chips for preclinical testing, mass spectrometry for postmarket monitoring, and detection/characterization of bacterial biofilm to prevent infections.
Collapse
Affiliation(s)
- Yi Wang
- Division of Biology, Chemistry, and Materials Science, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, Office of Medical Products and Tobacco, US Food and Drug Administration, Silver Spring, Maryland 20993, USA;
| | - Allan Guan
- Division of Biology, Chemistry, and Materials Science, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, Office of Medical Products and Tobacco, US Food and Drug Administration, Silver Spring, Maryland 20993, USA;
| | - Samanthi Wickramasekara
- Division of Biology, Chemistry, and Materials Science, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, Office of Medical Products and Tobacco, US Food and Drug Administration, Silver Spring, Maryland 20993, USA;
| | - K Scott Phillips
- Division of Biology, Chemistry, and Materials Science, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, Office of Medical Products and Tobacco, US Food and Drug Administration, Silver Spring, Maryland 20993, USA;
| |
Collapse
|
18
|
Tillander V, Alexson SEH, Cohen DE. Deactivating Fatty Acids: Acyl-CoA Thioesterase-Mediated Control of Lipid Metabolism. Trends Endocrinol Metab 2017; 28:473-484. [PMID: 28385385 PMCID: PMC5474144 DOI: 10.1016/j.tem.2017.03.001] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 03/01/2017] [Indexed: 12/28/2022]
Abstract
The cellular uptake of free fatty acids (FFA) is followed by esterification to coenzyme A (CoA), generating fatty acyl-CoAs that are substrates for oxidation or incorporation into complex lipids. Acyl-CoA thioesterases (ACOTs) constitute a family of enzymes that hydrolyze fatty acyl-CoAs to form FFA and CoA. Although biochemically and biophysically well characterized, the metabolic functions of these enzymes remain incompletely understood. Existing evidence suggests regulatory roles in controlling rates of peroxisomal and mitochondrial fatty acyl-CoA oxidation, as well as in the subcellular trafficking of fatty acids. Emerging data implicate ACOTs in the pathogenesis of metabolic diseases, suggesting that better understanding their pathobiology could reveal unique targets in the management of obesity, diabetes, and nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Veronika Tillander
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, 14186, Sweden
| | - Stefan E H Alexson
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, 14186, Sweden
| | - David E Cohen
- Division of Gastroenterology and Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA.
| |
Collapse
|