1
|
Yang L, Bhattacharya A, Peterson D, Li Y, Liu X, Marangoni E, Robila V, Zhang Y. Targeted dual degradation of HER2 and EGFR obliterates oncogenic signaling, overcomes therapy resistance, and inhibits metastatic lesions in HER2-positive breast cancer models. Drug Resist Updat 2024; 74:101078. [PMID: 38503142 PMCID: PMC11070302 DOI: 10.1016/j.drup.2024.101078] [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: 01/02/2024] [Revised: 02/29/2024] [Accepted: 03/08/2024] [Indexed: 03/21/2024]
Abstract
AIMS Human epidermal growth factor receptor 2 (HER2) is an oncogenic receptor tyrosine kinase amplified in approximately 20% of breast cancer (BC). HER2-targeted therapies are the linchpin of treating HER2-positive BC. However, drug resistance is common, and the main resistance mechanism is unknown. We tested the hypothesis that drug resistance results mainly from inadequate or lack of inhibition of HER2 and its family member epidermal growth factor receptor (EGFR). METHODS We used clinically relevant cell and tumor models to assess the impact of targeted degradation of HER2 and EGFR on trastuzumab resistance. Trastuzumab is the most common clinically used HER2 inhibitor. Targeted degradation of HER2 and EGFR was achieved using recombinant human protein PEPDG278D, which binds to the extracellular domains of the receptors. siRNA knockdown was used to assess the relative importance of EGFR and HER2 in trastuzumab resistance. RESULTS Both HER2 and EGFR are overexpressed in all trastuzumab-resistant HER2-positive BC cell and tumor models and that all trastuzumab-resistant models are highly vulnerable to targeted degradation of HER2 and EGFR. Degradation of HER2 and EGFR induced by PEPDG278D causes extensive inhibition of oncogenic signaling in trastuzumab-resistant HER2-positive BC cells. This is accompanied by strong growth inhibition of cultured cells, orthotopic patient-derived xenografts, and metastatic lesions in the brain and lung of trastuzumab-resistant HER2-positive BC. siRNA knockdown indicates that eliminating both HER2 and EGFR is necessary to maximize therapeutic outcome. CONCLUSIONS This study unravels the therapeutic vulnerability of trastuzumab-resistant HER2-positive BC and shows that an agent that targets the degradation of both HER2 and EGFR is highly effective in overcoming drug resistance in this disease. The findings provide new insights and innovations for advancing treatment of drug-resistant HER2-positive breast cancer that remains an unmet problem.
Collapse
Affiliation(s)
- Lu Yang
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Arup Bhattacharya
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Darrell Peterson
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, School of Pharmacy, Richmond, VA, USA
| | - Yun Li
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Xiaozhuo Liu
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | - Valentina Robila
- Department of Pathology, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Yuesheng Zhang
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
2
|
Liu YL, Zhu XX, Niu LY, Gao Y, Gan H, Wu ZN, Li J, Feng SX, Dou GF, Gu RL, Meng ZY. The Pharmacokinetics and Pharmacodynamics of A Novel Recombinant Activated Human Factor VII, GEN-0828, in Hemophilia B Mice. J Pharm Sci 2023; 112:877-883. [PMID: 36565924 DOI: 10.1016/j.xphs.2022.12.013] [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: 09/06/2022] [Revised: 12/16/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
GEN-0828, a proposed clinical candidate for hemophilia and trauma hemorrhage treatment, is a novel recombinant activated human factor VII (rFVIIa). The purpose of this paper is to compare the pharmacokinetics and pharmacodynamics of GEN-0828 in hemophilia B mice with those of NovoSeven®, the only marketed rFVIIa product worldwide., GEN-0828 and NovoSeven® showed similar affinity bioactivity to recombinant tissue factor (rTF) in vitro. Pharmacodynamics data indicated a generally similar hemostatic efficacy (ED50) of GEN-0828 (10.91 KIU·kg-1) and NovoSeven® (18.91 KIU·kg-1) at the doses studied in hemophilia B mice, while GEN-0828 represented a lower initial effective dosage compared with that of NovoSeven® in terms of both blood loss and APTT. GEN-0828 exhibited linear pharmacokinetic profiles in hemophilia B mice at the 30-338 KIU·kg-1 dose range, the comparative pharmacokinetic study with NovoSeven® indicated better characteristics than NovoSeven® in terms of the appropriate higher maximal concentration (Cmax) and area under the plasma concentration-time curve (AUClast) and longer mean residence time (MRT). In conclusion, GEN-0828 was a promising new type of rFVIIa compound with favourable pharmacokinetic and pharmacodynamic profiles.
Collapse
Affiliation(s)
- Yu-Lu Liu
- Beijing Institute of Radiation Medicine, Beijing 100850, China; College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Xiao-Xia Zhu
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Li-Yun Niu
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Ya Gao
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Hui Gan
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Zhuo-Na Wu
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Jian Li
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Su-Xiang Feng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of China, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| | - Gui-Fang Dou
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Ruo-Lan Gu
- Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Zhi-Yun Meng
- Beijing Institute of Radiation Medicine, Beijing 100850, China.
| |
Collapse
|
3
|
Yang L, Bhattacharya A, Li Y, Sexton S, Ling X, Li F, Zhang Y. Depleting receptor tyrosine kinases EGFR and HER2 overcomes resistance to EGFR inhibitors in colorectal cancer. J Exp Clin Cancer Res 2022; 41:184. [PMID: 35650607 PMCID: PMC9161494 DOI: 10.1186/s13046-022-02389-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/13/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) inhibitors, including cetuximab and panitumumab, are valuable therapeutics for colorectal cancer (CRC), but resistance to these inhibitors is common. The reason for such resistance is not well understood, which hampers development of better therapeutic strategies. Although activating mutations in KRAS, BRAF and PIK3CA are considered major drivers of CRC resistance to EGFR inhibitors, therapeutic targeting of these drug resistance drivers has not produced substantial clinical benefit. METHODS We exploited cell lines and mouse tumor models (cell line xenografts and patient derived xenografts) for experiments of genetic and pharmacologic depletion of EGFR and/or its family member HER2, including EGFR mutants, inhibition of EGFR ligand shedding, and biochemical analysis of signaling proteins, to delineate the mechanism of CRC resistance to EGFR inhibitors and to assess the therapeutic activity of PEPDG278D, which is a recombinant human protein that induces the degradation of both EGFR and HER2. RESULTS The sensitivity of CRC cells to cetuximab and panitumumab correlates with the ability of these drugs to induce EGFR downregulation. PEPDG278D strongly inhibits oncogenic signaling and growth of CRC cells by causing profound depletion of EGFR and HER2, regardless of activating mutations of KRAS, BRAF and PIK3CA. siRNA knockdown of EGFR or HER2 also inhibits CRC cells resistant to EGFR inhibitors. Tumors harboring mutated KRAS, BRAF and/or PIK3CA also overexpress EGFR ligands, further suggesting that EGFR signaling remains important to the tumors. While excessive tumor-generated high-affinity EGFR ligands block target engagement by PEPDG278D, aderbasib, an inhibitor of ADAM10 and ADAM17, enables PEPDG278D to exert strong antitumor activity by inhibiting ligand shedding. Moreover, adding fluorouracil, which is commonly used in CRC treatment, to the combination of PEPDG278D and aderbasib further enhances tumor inhibition. CONCLUSIONS Our study shows that CRC resistance to EGFR inhibitors results primarily from the inability of the inhibitors to downregulate their target and that a PEPDG278D-based combination treatment overcomes the resistance.
Collapse
Affiliation(s)
- Lu Yang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Department of Pharmacology and Toxicology, and Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - Arup Bhattacharya
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Yun Li
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Sandra Sexton
- Department of Animal Resources, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Xiang Ling
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Fengzhi Li
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Yuesheng Zhang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
- Department of Pharmacology and Toxicology, and Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA.
| |
Collapse
|
4
|
Anti-HK antibody reveals critical roles of a 20-residue HK region for Aβ-induced plasma contact system activation. Blood Adv 2022; 6:3090-3101. [PMID: 35147669 PMCID: PMC9131899 DOI: 10.1182/bloodadvances.2021006612] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 02/10/2022] [Indexed: 11/20/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder and the leading cause of dementia. Vascular abnormalities and neuroinflammation play roles in AD pathogenesis. Plasma contact activation, which leads to fibrin clot formation and bradykinin release, is elevated in many AD patients, likely due to the ability of AD's pathogenic peptide β-amyloid (Aβ) to induce its activation. Since overactivation of this system may be deleterious to AD patients, the development of inhibitors could be beneficial. Here, we show that 3E8, an antibody against a 20-amino acid region of high molecular weight kininogen's (HK) domain 6, inhibits Aβ-induced intrinsic coagulation. Mechanistically, 3E8 inhibits contact system activation by blocking the binding of prekallikrein (PK) and factor XI (FXI) to HK, thereby preventing their activation and the continued activation of factor XII (FXII). The 3E8 antibody can also disassemble HK/PK and HK/FXI complexes in normal human plasma in the absence of a contact system activator due to its strong binding affinity for HK, indicating its prophylactic ability. Furthermore, the binding of Aβ to both FXII and HK is critical for Aβ-mediated contact system activation. These results suggest that a 20-amino acid region of HK's domain 6 plays a critical role in Aβ-induced contact system activation, and this region may provide an effective strategy to inhibit or prevent contact system activation in related disorders.
Collapse
|
5
|
Hu YN, Gan YM, Zhang YP, Ruan DD, Zhu YB, Lin XF, Fang ZT, Liao LS, Tang FQ, Luo JW. A novel compound heterozygous variant linked to hematuria in a family with hereditary factor VII deficiency. J Gene Med 2021; 24:e3398. [PMID: 34786791 DOI: 10.1002/jgm.3398] [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: 05/06/2021] [Revised: 09/06/2021] [Accepted: 09/28/2021] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Hereditary Factor VII Deficiency (FVIID) is a rare congenital autosomal recessive bleeding disorder. In clinical manifestations, its onset is caused by variant of the F7 gene (NM_019616) with strong heterogeneity. We identified a family with hematuria caused by a novel F7 compound heterozygous variant and studied the FVIID-dependent mechanism impacted by these variants. METHODS Coagulation factors in the proband were functionally verified. We located pathogenic variants in relevant genes using next-generation sequencing after target enrichment and verified them by Sanger sequencing. We examined the coagulation activity and secretion pattern of recombinant FVII variants expressed in cells and observed their location and stability by immunofluorescence. RESULTS We found a missense variant c.1207G>A (p.Gly403Ser) and a frameshift variant c.154_155del (p.Arg53fs) in the F7 gene of the proband. FVII activity tests showed that the variants significantly decreased its presence in the cell culture supernatant. Moreover, the R53fs mutant lacked the FVII functional domain and had no detectable activity. Immunofluorescence indicated that the p.Gly403Ser variant was distributed to the cell membrane and cytoplasm, while the FVII R53fs variant was not detected. Deficient FVII protein function and severe coagulation disorder are the likely causes of hematuria and other bleeding symptoms in the proband. CONCLUSION The newly discovered F7 gene variants enrich the spectrum of hereditary FVII deficiency and provide a new foundation for the diagnosis and treatment of this type of coagulation disorder.
Collapse
Affiliation(s)
- Ya-Nan Hu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China.,Fujian provincial hospital, Fuzhou, China
| | - Yu-Mian Gan
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China.,Fujian provincial hospital, Fuzhou, China
| | - Yan-Ping Zhang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China.,Fujian provincial hospital, Fuzhou, China
| | - Dan-Dan Ruan
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China.,Fujian provincial hospital, Fuzhou, China
| | - Yao-Bin Zhu
- Department of Traditional Chinese Medicine, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xin-Fu Lin
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China.,Fujian provincial hospital, Fuzhou, China
| | - Zhu-Ting Fang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China.,Fujian provincial hospital, Fuzhou, China
| | - Li-Sheng Liao
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China.,Fujian provincial hospital, Fuzhou, China
| | - Fa-Qiang Tang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China.,Fujian provincial hospital, Fuzhou, China
| | - Jie-Wei Luo
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China.,Fujian provincial hospital, Fuzhou, China
| |
Collapse
|
6
|
Frey AM, Chaput D, Shaw LN. Insight into the human pathodegradome of the V8 protease from Staphylococcus aureus. Cell Rep 2021; 35:108930. [PMID: 33826899 PMCID: PMC8054439 DOI: 10.1016/j.celrep.2021.108930] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/03/2020] [Accepted: 03/11/2021] [Indexed: 12/03/2022] Open
Abstract
Staphylococcus aureus possesses ten extracellular proteases with mostly unknown targets in the human proteome. To assist with bacterial protease target discovery, we have applied and compared two N-terminomics methods to investigate cleavage of human serum proteins by S. aureus V8 protease, discovering 85 host-protein targets. Among these are virulence-relevant complement, iron sequestration, clotting cascade, and host protease inhibitor proteins. Protein cleavage sites have been identified, providing insight into the disruption of host protein function by V8. Complement proteins are cleaved within peptidase and sushi domains, and host protease inhibitors are cleaved outside their protease-trapping motifs. Our data highlight the potential for further application of N-terminomics in discovery of bacterial protease substrates in other host niches and provide omics-scale insight into the role of the V8 protease in S. aureus pathogenesis. S. aureus-secreted proteases are central to disease causation, but the discovery of their host substrates has been limited. Frey et al. use N-terminomic approaches to uncover human serum targets of the V8 protease that are from virulence-relevant processes such as the host inflammatory network and nutrient sequestration.
Collapse
Affiliation(s)
- Andrew Michael Frey
- Department of Cell Biology, Microbiology & Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Dale Chaput
- Department of Cell Biology, Microbiology & Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Lindsey Neil Shaw
- Department of Cell Biology, Microbiology & Molecular Biology, University of South Florida, Tampa, FL 33620, USA.
| |
Collapse
|
7
|
Peoples N, Strang C. Complement Activation in the Central Nervous System: A Biophysical Model for Immune Dysregulation in the Disease State. Front Mol Neurosci 2021; 14:620090. [PMID: 33746710 PMCID: PMC7969890 DOI: 10.3389/fnmol.2021.620090] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/12/2021] [Indexed: 01/08/2023] Open
Abstract
Complement, a feature of the innate immune system that targets pathogens for phagocytic clearance and promotes inflammation, is tightly regulated to prevent damage to host tissue. This regulation is paramount in the central nervous system (CNS) since complement proteins degrade neuronal synapses during development, homeostasis, and neurodegeneration. We propose that dysregulated complement, particularly C1 or C3b, may errantly target synapses for immune-mediated clearance, therefore highlighting regulatory failure as a major potential mediator of neurological disease. First, we explore the mechanics of molecular neuroimmune relationships for the regulatory proteins: Complement Receptor 1, C1-Inhibitor, Factor H, and the CUB-sushi multiple domain family. We propose that biophysical and chemical principles offer clues for understanding mechanisms of dysregulation. Second, we describe anticipated effects to CNS disease processes (particularly Alzheimer's Disease) and nest our ideas within existing basic science, clinical, and epidemiological findings. Finally, we illustrate how the concepts presented within this manuscript provoke new ways of approaching age-old neurodegenerative processes. Every component of this model is testable by straightforward experimentation and highlights the untapped potential of complement dysregulation as a driver of CNS disease. This includes a putative role for complement-based neurotherapeutic agents and companion biomarkers.
Collapse
|
8
|
Zhang Y. The root cause of drug resistance in HER2-positive breast cancer and the therapeutic approaches to overcoming the resistance. Pharmacol Ther 2020; 218:107677. [PMID: 32898548 DOI: 10.1016/j.pharmthera.2020.107677] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 08/28/2020] [Indexed: 12/19/2022]
Abstract
HER2 is a well-known oncogenic receptor tyrosine kinase. HER2 gene amplification occurs in about 20% of breast cancer (BC), which leads to overexpression of HER2 protein, known as HER2-positive BC. Inhibitors of HER2 have significantly improved the prognosis of patients with this subset of BC. Since 1998, seven HER2 inhibitors have been developed to treat this disease. However, drug resistance is common and remains a major unresolved clinical problem. Patients typically show disease progression after some time on treatment. This review discusses the complexity and diversified nature of HER2 signaling, the mechanisms of actions and therapeutic activities of all HER2 inhibitors, the roles of HER2 and other signaling proteins in HER2-positive BC resistant to the inhibitors, the non-cell-autonomous mechanisms of drug resistance, and the heterogeneity of tumor HER2 expression. The review presents the concept that drug resistance in HER2-positive BC results primarily from the inability of HER2 inhibitors to deplete HER2. Emerging therapeutics that are promising for overcoming drug resistance are also discussed.
Collapse
Affiliation(s)
- Yuesheng Zhang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, USA.
| |
Collapse
|
9
|
Begic E, Hadzidedic S, Obradovic S, Begic Z, Causevic M. Increased Levels of Coagulation Factor XI in Plasma Are Related to Alzheimer's Disease Diagnosis. J Alzheimers Dis 2020; 77:375-386. [PMID: 32804133 DOI: 10.3233/jad-200358] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Alzheimer's disease is a complex disorder of unclear etiology that develops in the elderly population. It is a debilitating, progressive neurodegeneration for which disease-modifying therapies do not exist. Previous studies have suggested that, for a subset of patients, dysregulation in hemostasis might be one of the molecular mechanisms that ultimately leads to the development of neurodegeneration resulting in cognitive decline that represents the most prominent symptomatic characteristic of Alzheimer's disease. OBJECTIVE To examine a relationship between factors that are part of coagulation and anticoagulation pathways with cognitive decline that develops during Alzheimer's disease. METHODS SOMAscan assay was used to measure levels of coagulation/anticoagulation factors V, VII, IX, X, Xa, XI, antithrombin III, protein S, protein C, and activated protein C in plasma samples obtained from three groups of subjects: 1) subjects with stable cognitively healthy function, 2) subjects with stable mild cognitive impairment, and 3) subjects diagnosed with probable Alzheimer's disease. RESULTS Our results show that protein levels of coagulation factor XI are significantly increased in patients who are diagnosed with probable Alzheimer's disease compared with cognitively healthy subjects or patients diagnosed with mild cognitive impairment. Furthermore, our results demonstrate that significant predictors of Alzheimer's-type diagnosis are factors IX and XI-an increase in both factors is associated with a reduction in cognitive function. CONCLUSION Our study justifies further investigations of biological pathways involving coagulation/anticoagulation factors in relation to dementia, including dementia resulting from Alzheimer's-type neurodegeneration.
Collapse
Affiliation(s)
- Edin Begic
- Department of Pharmacology, Sarajevo Medical School, Sarajevo School of Science and Technology, Sarajevo, Bosnia and Herzegovina.,Department of Cardiology, General Hospital "Prim.Dr. Abdulah Nakas", Sarajevo, Bosnia and Herzegovina
| | - Suncica Hadzidedic
- Computer Science and Information Systems Department, Sarajevo School of Science and Technology, Sarajevo, Bosnia and Herzegovina.,Department of Computer Science, Durham University, Durham, UK
| | - Slobodan Obradovic
- Clinic for Emergency and Internal Medicine, Military Medical Academy, University of Defense, Belgrade, Serbia
| | - Zijo Begic
- Department of Pediatric Cardiology, Pediatric Clinic, Clinical Center University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Mirsada Causevic
- Department of Pharmacology, Sarajevo Medical School, Sarajevo School of Science and Technology, Sarajevo, Bosnia and Herzegovina
| |
Collapse
|
10
|
Current Understanding of the Emerging Role of Prolidase in Cellular Metabolism. Int J Mol Sci 2020; 21:ijms21165906. [PMID: 32824561 PMCID: PMC7460564 DOI: 10.3390/ijms21165906] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/11/2020] [Accepted: 08/15/2020] [Indexed: 12/12/2022] Open
Abstract
Prolidase [EC 3.4.13.9], known as PEPD, cleaves di- and tripeptides containing carboxyl-terminal proline or hydroxyproline. For decades, prolidase has been thoroughly investigated, and several mechanisms regulating its activity are known, including the activation of the β1-integrin receptor, insulin-like growth factor 1 receptor (IGF-1) receptor, and transforming growth factor (TGF)-β1 receptor. This process may result in increased availability of proline in the mitochondrial proline cycle, thus making proline serve as a substrate for the resynthesis of collagen, an intracellular signaling molecule. However, as a ligand, PEPD can bind directly to the epidermal growth factor receptor (EGFR, epidermal growth factor receptor 2 (HER2)) and regulate cellular metabolism. Recent reports have indicated that PEPD protects p53 from uncontrolled p53 subcellular activation and its translocation between cellular compartments. PEPD also participates in the maturation of the interferon α/β receptor by regulating its expression. In addition to the biological effects, prolidase demonstrates clinical significance reflected in the disease known as prolidase deficiency. It is also known that prolidase activity is affected in collagen metabolism disorders, metabolic, and oncological conditions. In this article, we review the latest knowledge about prolidase and highlight its biological function, and thus provide an in-depth understanding of prolidase as a dipeptidase and protein regulating the function of key biomolecules in cellular metabolism.
Collapse
|
11
|
Yang L, Li Y, Bhattacharya A, Zhang Y. A recombinant human protein targeting HER2 overcomes drug resistance in HER2-positive breast cancer. Sci Transl Med 2020; 11:11/476/eaav1620. [PMID: 30674653 DOI: 10.1126/scitranslmed.aav1620] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 12/05/2018] [Indexed: 12/17/2022]
Abstract
Resistance to current human epidermal growth factor receptor 2 (HER2) inhibitors, such as trastuzumab (Ttzm), is a major unresolved clinical problem in HER2-positive breast cancer (HER2-BC). Because HER2 remains overexpressed in drug-resistant HER2-BC cells, we investigated whether PEPDG278D can overcome the resistance. PEPDG278D is a recombinant enzymatically inactive mutant of human peptidase D, which strongly inhibits HER2 in cancer cells by binding to its extracellular domain. Here, we show that PEPDG278D is highly active in preclinical models of HER2-BC resistant to Ttzm and other HER2 inhibitors and also enhances the therapeutic efficacy of paclitaxel. The therapeutic activity is underscored by its ability to bind to HER2 and free it from protection by mucin 4, disrupt its interplay with other receptor tyrosine kinases, and subsequently direct HER2 for degradation. PEPDG278D also down-regulates epidermal growth factor receptor, which contributes to drug resistance in HER2-BC. In contrast, Ttzm, whose therapeutic activity also depends on its binding to the extracellular domain of HER2, cannot perform any of these functions of PEPDG278D PEPDG278D inhibits HER2-BC cells and tumors that carry clinically relevant molecular changes that confer resistance to Ttzm. Our results show that HER2 remains a critical target in drug-resistant HER2-BC and that PEPDG278D is a promising agent for overcoming drug resistance in this disease.
Collapse
Affiliation(s)
- Lu Yang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Yun Li
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.,Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Arup Bhattacharya
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Yuesheng Zhang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA. .,Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
12
|
Rajan MR, Sotak M, Barrenäs F, Shen T, Borkowski K, Ashton NJ, Biörserud C, Lindahl TL, Ramström S, Schöll M, Lindahl P, Fiehn O, Newman JW, Perkins R, Wallenius V, Lange S, Börgeson E. Comparative analysis of obesity-related cardiometabolic and renal biomarkers in human plasma and serum. Sci Rep 2019; 9:15385. [PMID: 31659186 PMCID: PMC6817872 DOI: 10.1038/s41598-019-51673-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 10/02/2019] [Indexed: 12/19/2022] Open
Abstract
The search for biomarkers associated with obesity-related diseases is ongoing, but it is not clear whether plasma and serum can be used interchangeably in this process. Here we used high-throughput screening to analyze 358 proteins and 76 lipids, selected because of their relevance to obesity-associated diseases, in plasma and serum from age- and sex-matched lean and obese humans. Most of the proteins/lipids had similar concentrations in plasma and serum, but a subset showed significant differences. Notably, a key marker of cardiovascular disease PAI-1 showed a difference in concentration between the obese and lean groups only in plasma. Furthermore, some biomarkers showed poor correlations between plasma and serum, including PCSK9, an important regulator of cholesterol homeostasis. Collectively, our results show that the choice of biofluid may impact study outcome when screening for obesity-related biomarkers and we identify several markers where this will be the case.
Collapse
Affiliation(s)
- Meenu Rohini Rajan
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Matus Sotak
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Barrenäs
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Cell & Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Tong Shen
- NIH West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, USA
| | - Kamil Borkowski
- NIH West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, USA
| | - Nicholas J Ashton
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Physiology and Neuroscience, University of Gothenburg, Gothenburg, Sweden
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Christina Biörserud
- Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Tomas L Lindahl
- Department of Clinical Chemistry and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Sofia Ramström
- Department of Clinical Chemistry and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
- Cardiovascular Research Centre, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Physiology and Neuroscience, University of Gothenburg, Gothenburg, Sweden
- Dementia Research Centre, Institute of Neurology, University College London, London, UK
| | - Per Lindahl
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Oliver Fiehn
- NIH West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, USA
| | - John W Newman
- NIH West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, USA
- Department of Nutrition, University of California Davis, Davis, USA
- USDA, ARS, Western Human Nutrition Research Center, Davis, USA
| | - Rosie Perkins
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Ville Wallenius
- Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Stephan Lange
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
- Division of Cardiology, School of Medicine, University of California San Diego, San Diego, USA
| | - Emma Börgeson
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
- Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
13
|
Shahbazi S, Mahdian R. Factor VII Gene Defects: Review of Functional Studies and Their Clinical Implications. IRANIAN BIOMEDICAL JOURNAL 2019; 23. [PMID: 30797223 PMCID: PMC6462297 DOI: 10.29252/.23.3.165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Coagulation factors belong to a family of plasma glycosylated proteins that should be activated for appropriate blood coagulation. Congenital deficiencies of these factors cause inheritable hemorrhagic diseases. Factor VII (FVII) deficiency is a rare bleeding disorder with variable clinical symptoms. Various mutations have been identified throughout the F7 gene and can affect all the protein domains. The results of previous experiments have partly revealed the correlation between genotype and phenotype in patients with FVII deficiency. Nevertheless, each particular variant may affect the coagulative function of FVII, mainly via altering its expression level, extra-cellular secretion, tissue factor binding affinity, or proteolytic activity. The pathogenicity of the variants and molecular mechanisms responsible for clinical symptoms in patients with FVII deficiency should be characterized via in silico and in vitro, as well as in vivo functional studies. This review has highlighted the most important functional studies reported on F7 gene variants, including relevant reports regarding Iranian FVII deficiency patients.
Collapse
Affiliation(s)
- Shirin Shahbazi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Reza Mahdian
- Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran,Corresponding Author: Reza Mahdian Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran; Tel.& Fax: (+98-21) 64112439; Mobile: (+98-912)7988388; E-mail:
| |
Collapse
|
14
|
Abstract
The fundamental pathology in Alzheimer's disease (AD) is neuronal dysfunction leading to cognitive impairment. The amyloid-β peptide (Aβ), derived from amyloid precursor protein, is one driver of AD, but how it leads to neuronal dysfunction is not established. In this Review, I discuss the complexity of AD and possible cause-and-effect relationships between Aβ and the vascular and hemostatic systems. AD can be considered a multifactorial syndrome with various contributing pathological mechanisms. Therefore, as is routinely done with cancer, it will be important to classify patients with respect to their disease signature so that specific pathologies, including vascular pathways, can be therapeutically targeted.
Collapse
|
15
|
Yang L, Li Y, Bhattacharya A, Zhang Y. Dual inhibition of ErbB1 and ErbB2 in cancer by recombinant human prolidase mutant hPEPD-G278D. Oncotarget 2018; 7:42340-42352. [PMID: 27286447 PMCID: PMC5173139 DOI: 10.18632/oncotarget.9851] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 05/20/2016] [Indexed: 01/14/2023] Open
Abstract
ErbB1 and ErbB2 are oncogenic cell surface receptor tyrosine kinases, linked to many forms of human cancer, and are major cancer therapeutic targets. Many lines of evidence indicate that targeting ErbB1 and ErbB2 is an important cancer therapeutic approach. We recently found that a recombinant enzymatically-inactive mutant of human prolidase, i.e., hPEPD-G278D, is an inhibitory ligand of ErbB2 and strongly inhibits ErbB2-overexpressing cells in vitro and in vivo. hPEPD-G278D also binds to ErbB1. Here, we show that hPEPD-G278D binds to ErbB1 with high affinity, initially activating ErbB1 but later silencing it, and that deletion of subdomain 2 in ErbB1 extracellular domain abolishes the binding. The proliferation of ErbB1-overexpressing cells is strongly inhibited by hPEPD-G278D, regardless of ErbB2 expression or cell type, whereas cells lacking ErbB1 and ErbB2 are insensitive to it. In contrast, EGF, another ErbB1 ligand, either stimulates or mildly inhibits cell proliferation. Moreover, hPEPD-G278D treatment of mice bearing ErbB1-overexpressing tumors leads to tumor regression, which is accompanied by down regulation and decreased phosphorylation of ErbB1 and ErbB2 as well as decreased phosphorylation of downstream signaling molecules and activation of apoptosis in the tumor tissues. We conclude that hPEPD-G278D is a dual inhibitor of ErbB1 and ErbB2 and selectively targets cancer cells overexpressing ErbB1 and/or ErbB2. Moreover, our finding that both receptors are silenced in cancer cells by hPEPD-G278D highlights an unusual consequence of ligand-receptor interaction.
Collapse
Affiliation(s)
- Lu Yang
- Departments of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Yun Li
- Departments of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA.,Urology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Arup Bhattacharya
- Departments of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Yuesheng Zhang
- Departments of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA.,Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY, USA
| |
Collapse
|
16
|
Ahn HJ, Chen ZL, Zamolodchikov D, Norris EH, Strickland S. Interactions of β-amyloid peptide with fibrinogen and coagulation factor XII may contribute to Alzheimer's disease. Curr Opin Hematol 2018; 24:427-431. [PMID: 28661939 DOI: 10.1097/moh.0000000000000368] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE OF REVIEW To review the evidence that the Alzheimer peptide β-amyloid interacts with the blood coagulation system and influences the pathophysiology of the disease. RECENT FINDINGS β-amyloid can interact with fibrinogen and blood coagulation factor XII and trigger ischemia and inflammation. SUMMARY β-amyloid interacts with fibrinogen and factor XII. These interactions can lead to increased clotting, abnormal clot formation, persistent fibrin deposition, and generation of proinflammatory molecules. These events can damage neurons and could contribute to the cognitive decline in Alzheimer's disease patients.
Collapse
Affiliation(s)
- Hyung J Ahn
- aPatricia and John Rosenwald Laboratory of Neurobiology and Genetics, Rockefeller University, New York City bRegeneron Pharmaceuticals, Tarrytown, New York, USA *Hyung J. Ahn, Zu-Lin Chen, and Daria Zamolodchikov contributed equally to this article
| | | | | | | | | |
Collapse
|
17
|
Anticoagulants inhibit proteolytic clearance of plasma amyloid beta. Oncotarget 2017; 9:5614-5626. [PMID: 29464022 PMCID: PMC5814162 DOI: 10.18632/oncotarget.23718] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 12/22/2017] [Indexed: 01/30/2023] Open
Abstract
We recently discovered a plasma proteolysis pathway, termed the FXII-FVII pathway which is composed of coagulation proteases, and found it to be mainly responsible for the clearance of Aβ42 in the plasma in mice. Aβ42 and Aβ40 are the main Aβ forms in Alzheimer’s disease (AD). In the present study, in vitro assays, wild type (WT) mice and J20 mice (a transgenic AD model) are used to assess the degradation of Aβ40 and Aβ42 by the FXII-FVII pathway and the impact of anticoagulants on such degradation. Four clinically available and mechanistically distinct anticoagulants are evaluated, including dabigatran, enoxaparin (EP), rivaroxaban and warfarin. Each anticoagulant significantly elevates plasma level of synthetic Aβ42 in WT mice, among which EP is the most effective. The differential efficacies of the anticoagulants in elevating plasma Aβ42 level match closely with their inhibitory mechanisms towards the FXII-FVII pathway. Plasma Aβ40 is also degraded by the FXII-FVII pathway and is protected by EP. Moreover, the FXII-FVII pathway is significantly activated in J20 mice, but EP inhibits the activation and significantly elevates plasma levels of both Aβ40 and Aβ42. Taken together, our results shed new light on Aβ metabolism, reveal a novel function of anticoagulants, and suggest a novel approach to potentially developing plasma Aβ as an AD biomarker.
Collapse
|
18
|
Abstract
p53 tumor suppressor responds to various cellular stresses and regulates cell fate. Here, we show that peptidase D (PEPD) binds and suppresses over half of nuclear and cytoplasmic p53 under normal conditions, independent of its enzymatic activity. Eliminating PEPD causes cell death and tumor regression due to p53 activation. PEPD binds to the proline-rich domain in p53, which inhibits phosphorylation of nuclear p53 and MDM2-mediated mitochondrial translocation of nuclear and cytoplasmic p53. However, the PEPD-p53 complex is critical for p53 response to stress, as stress signals doxorubicin and H2O2 each must free p53 from PEPD in order to achieve robust p53 activation, which is mediated by reactive oxygen species. Thus, PEPD stores p53 for the stress response, but this also renders cells dependent on PEPD for survival, as it suppresses p53. This finding provides further understanding of p53 regulation and may have significant implications for the treatment of cancer and other diseases. p53 is a pivotal tumour suppressor that is activated by various cellular stress inducers. Here, the authors show that peptidase D (PEPD) promotes the growth of cancer cells by suppressing p53 and that the complex PEPD-p53 is critical for robust p53 activation in response to stress signals.
Collapse
|