1
|
Wang L, Deng X, Tang J, Gong Y, Bu S, Li Z, Liao B, Ding Y, Dai T, Liao Y, Li Y. Identification of EARS2 as a Potential Biomarker with Diagnostic, Prognostic, and Therapeutic Implications in Colorectal Cancer. Immunotargets Ther 2025; 14:65-85. [PMID: 39906030 PMCID: PMC11791672 DOI: 10.2147/itt.s499680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/18/2025] [Indexed: 02/06/2025] Open
Abstract
Purpose Colorectal cancer (CRC) is a prevalent malignancy, and lactate metabolism significantly influences tumorigenesis and progression. This study identifies key genes associated with lactic acid metabolism and explore their impact on CRC. Patients and Methods This study utilized data from The Cancer Genome Atlas, Gene Expression Omnibus, other public databases, and our institutional resources. Machine learning identified key lactate metabolism-related genes. Receiver Operating Characteristic analysis, Kaplan-Meier analysis, and the construction of a nomogram model were conducted to assess the diagnostic and prognostic significance of the key lactate metabolism-related gene EARS2. EARS2 expression in colorectal tissue was validated using both publicly available external data and samples from our institution. To investigate the mechanisms underlying EARS2 in CRC, Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, Gene Set Enrichment Analysis, and Protein-Protein Interaction analyses were performed, alongside the construction of miRNA-mRNA interaction networks. Additionally, the relationships between EARS2 and immune cell infiltration, as well as responses to drug therapy, were examined. Following the knockdown of EARS2, we assessed cell proliferation, migration capabilities, and apoptosis. Statistical analyses were conducted using R and GraphPad Prism software. Results ERAS2 was overexpressed in CRC tissues compared to normal and adenoma tissues, with higher expression levels correlating with aberrant lactate metabolism and poorer patient prognosis. EARS2 was involved in pathways such as neuroactive ligand-receptor interactions, protein digestion, and cholesterol metabolism, and it was associated with immune cell infiltration and responses to drug treatment. Additionally, the knockdown of EARS2 inhibited the proliferation, migration, and invasion of CRC cells while enhancing their apoptosis. Conclusion Elevated expression of EARS2 is associated with abnormal lactate metabolism, immune cell infiltration, altered therapeutic sensitivity, and poorer survival outcomes in CRC. This correlation suggests that EARS2 may serve as a potential target for the diagnosis, prognosis, and therapeutic intervention in CRC.
Collapse
Affiliation(s)
- Ling Wang
- Department of Infectious Diseases, Second Affiliated Hospital, Chongqing Medical University, Chongqing, People’s Republic of China
- Department of Phase I Clinical Trial Ward, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, People’s Republic of China
| | - Xuemei Deng
- Department of Infectious Diseases, Second Affiliated Hospital, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Jiaxi Tang
- Department of Anesthesiology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yi Gong
- Department of Phase I Clinical Trial Ward, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, People’s Republic of China
| | - Shaojin Bu
- Department of Anesthesiology, Fengdu People’s Hospital, Chongqing, People’s Republic of China
| | - Zuli Li
- Department of Infectious Diseases, Second Affiliated Hospital, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Bin Liao
- Department of Phase I Clinical Trial Ward, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, People’s Republic of China
| | - Yao Ding
- Department of Phase I Clinical Trial Ward, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, People’s Republic of China
| | - Tingting Dai
- Department of Infectious Diseases, Second Affiliated Hospital, Chongqing Medical University, Chongqing, People’s Republic of China
- Department of Medical Oncology, Chongqing University Fuling Hospital, Chongqing, China
| | - Yong Liao
- Department of Infectious Diseases, Second Affiliated Hospital, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Yongsheng Li
- Department of Phase I Clinical Trial Ward, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, People’s Republic of China
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, People’s Republic of China
| |
Collapse
|
2
|
Dong Y, Yin L, Huang J, Hu D, Sun J, Zhang Z, Li Z, Zhong BY, Zhu R, Wang G. 99mTc/ 90Y radiolabeled biodegradable gel microspheres for lung shutting fraction assessment and radioembolization in hepatocellular carcinoma theranostics. Mater Today Bio 2024; 29:101367. [PMID: 39687798 PMCID: PMC11647226 DOI: 10.1016/j.mtbio.2024.101367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/13/2024] [Accepted: 11/23/2024] [Indexed: 12/18/2024] Open
Abstract
Transarterial radioembolization (TARE) is a well-established clinical therapy for the treatment of patients with intermediate to advanced hepatocellular carcinoma (HCC) or those who are ineligible for radical treatment. However, commercialized radioactive microspheres still have some issues, such as high density, complicated preparation, non-biodegradability. Furthermore, the use of different radioactive microspheres during TARE and lung shunt fraction assessment has led to inconsistencies in biodistribution in certain cases. This study employed biodegradable hyaluronic acid (HA) as the backbone and modified with bisphosphonate and methacrylic acid to prepare biodegradable gel microspheres (HAMS) using the water-in-oil emulsification and photo-crosslinking for labeling the diagnostic radionuclide of 99mTc and therapeutic radionuclide of 90Y. Both 99mTc radiolabeled HAMS (99mTc-HAMS) and radiolabeled 90Y-HAMS (90Y-HAMS) were highly efficient in radiolabeling and exhibited excellent radiostability in vitro and in vivo. 99mTc-HAMS are highly effective in assessing the LSF, while 90Y-HAMS, administered though TARE, are effective in inhibiting the growth of in situ HCC without any side effects. Both 99mTc-HAMS and 90Y-HAMS have promising clinical applications in HCC theranostics.
Collapse
Affiliation(s)
- Yi Dong
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Lingling Yin
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Jintao Huang
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Di Hu
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Jing Sun
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Zhe Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Zhihao Li
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Bin-Yan Zhong
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Ran Zhu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Guanglin Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| |
Collapse
|
3
|
Fink SP, Triggs-Raine B. Genetic Deficiencies of Hyaluronan Degradation. Cells 2024; 13:1203. [PMID: 39056785 PMCID: PMC11275217 DOI: 10.3390/cells13141203] [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: 05/30/2024] [Revised: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Hyaluronan (HA) is a large polysaccharide that is broadly distributed and highly abundant in the soft connective tissues and embryos of vertebrates. The constitutive turnover of HA is very high, estimated at 5 g per day in an average (70 kg) adult human, but HA turnover must also be tightly regulated in some processes. Six genes encoding homologues to bee venom hyaluronidase (HYAL1, HYAL2, HYAL3, HYAL4, HYAL6P/HYALP1, SPAM1/PH20), as well as genes encoding two unrelated G8-domain-containing proteins demonstrated to be involved in HA degradation (CEMIP/KIAA1199, CEMIP2/TMEM2), have been identified in humans. Of these, only deficiencies in HYAL1, HYAL2, HYAL3 and CEMIP have been identified as the cause or putative cause of human genetic disorders. The phenotypes of these disorders have been vital in determining the biological roles of these enzymes but there is much that is still not understood. Deficiencies in these HA-degrading proteins have been created in mice and/or other model organisms where phenotypes could be analyzed and probed to expand our understanding of HA degradation and function. This review will describe what has been found in human and animal models of hyaluronidase deficiency and discuss how this has advanced our understanding of HA's role in health and disease.
Collapse
Affiliation(s)
- Stephen P. Fink
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - Barbara Triggs-Raine
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Children’s Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
| |
Collapse
|
4
|
Murai T. Transmembrane signaling through single-spanning receptors modulated by phase separation at the cell surface. Eur J Cell Biol 2024; 103:151413. [PMID: 38631097 DOI: 10.1016/j.ejcb.2024.151413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/19/2024] Open
Abstract
A wide variety of transmembrane signals are transduced by cell-surface receptors that activate intracellular signaling molecules. In particular, receptor clustering in the plasma membrane plays a critical role in these processes. Single-spanning or single-pass transmembrane proteins are among the most significant types of membrane receptors, which include adhesion receptors, such as integrins, CD44, cadherins, and receptor tyrosine kinases. Elucidating the molecular mechanisms underlying the regulation of the activity of these receptors is of great significance. Liquid-liquid phase separation (LLPS) is a recently emerging paradigm in cellular physiology for the ubiquitous regulation of the spatiotemporal dynamics of various signaling pathways. This study describes the emerging features of transmembrane signaling through single-spanning receptors from the perspective of phase separation. Possible physicochemical modulations of LLPS-based transmembrane signaling are also discussed.
Collapse
Affiliation(s)
- Toshiyuki Murai
- Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
5
|
Stock C. pH-regulated single cell migration. Pflugers Arch 2024; 476:639-658. [PMID: 38214759 PMCID: PMC11006768 DOI: 10.1007/s00424-024-02907-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/21/2023] [Accepted: 01/02/2024] [Indexed: 01/13/2024]
Abstract
Over the last two decades, extra- and intracellular pH have emerged as fundamental regulators of cell motility. Fundamental physiological and pathological processes relying on appropriate cell migration, such as embryonic development, wound healing, and a proper immune defense on the one hand, and autoimmune diseases, metastatic cancer, and the progression of certain parasitic diseases on the other, depend on surrounding pH. In addition, migrating single cells create their own localized pH nanodomains at their surface and in the cytosol. By this means, the migrating cells locally modulate their adhesion to, and the re-arrangement and digestion of, the extracellular matrix. At the same time, the cytosolic nanodomains tune cytoskeletal dynamics along the direction of movement resulting in concerted lamellipodia protrusion and rear end retraction. Extracellular pH gradients as found in wounds, inflamed tissues, or the periphery of tumors stimulate directed cell migration, and long-term exposure to acidic conditions can engender a more migratory and invasive phenotype persisting for hours up to several generations of cells after they have left the acidic milieu. In the present review, the different variants of pH-dependent single cell migration are described. The underlying pH-dependent molecular mechanisms such as conformational changes of adhesion molecules, matrix protease activity, actin (de-)polymerization, and signaling events are explained, and molecular pH sensors stimulated by H+ signaling are presented.
Collapse
Affiliation(s)
- Christian Stock
- Department of Gastroenterology, Hepatology, Infectiology & Endocrinology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
6
|
Tafech A, Stéphanou A. On the Importance of Acidity in Cancer Cells and Therapy. BIOLOGY 2024; 13:225. [PMID: 38666837 PMCID: PMC11048434 DOI: 10.3390/biology13040225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024]
Abstract
Cancer cells are associated with high glycolytic activity, which results in acidification of the tumor microenvironment. The occurrence of this stressful condition fosters tumor aggressiveness, with the outcome of invasiveness and metastasis that are linked to a poor clinical prognosis. Acidosis can be both the cause or consequence of alterations in the functions and expressions of transporters involved in intracellular acidity regulation. This review aims to explore the origin of acidity in cancer cells and the various mechanisms existing in tumors to resist, survive, or thrive in the acidic environment. It highlights the difficulties in measuring the intracellular pH evolution that impedes our understanding of the many regulatory and feedback mechanisms. It finally presents the consequences of acidity on tumor development as well as the friend or foe role of acidity in therapy.
Collapse
Affiliation(s)
| | - Angélique Stéphanou
- Université Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, TIMC, 38000 Grenoble, France
| |
Collapse
|
7
|
Shinohara Y, Komiya Y, Morimoto K, Endo Y, Terashima M, Suzuki T, Takino T, Ninomiya I, Yamada H, Uto Y. Development of UTX-143, a selective sodium-hydrogen exchange subtype 5 inhibitor, using amiloride as a lead compound. Bioorg Med Chem 2024; 99:117603. [PMID: 38246115 DOI: 10.1016/j.bmc.2024.117603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/06/2024] [Accepted: 01/11/2024] [Indexed: 01/23/2024]
Abstract
NHE5, an isoform of the Na+/H+ exchanger (NHE) protein, is an ion-transporting membrane protein that regulates intracellular pH and is highly expressed in colorectal adenocarcinoma. Therefore, we hypothesized that NHE5 inhibitors can be used as anticancer drugs. However, because NHE1 is ubiquitously expressed in all cells, it is extremely important to demonstrate its selective inhibitory activity against NHE5. We used amiloride, an NHE non-selective inhibitor, as a lead compound and created UTX-143, which has NHE5-selective inhibitory activity, using a structure-activity relationship approach. UTX-143 showed selective cytotoxic effects on cancer cells and reduced the migratory and invasive abilities of cancer cells. These results suggest a new concept wherein drugs exhibit cancer-specific cytotoxic effects through selective inhibition of NHE5 and the possibility of UTX-143 as a lead NHE5-selective inhibitor.
Collapse
Affiliation(s)
- Yusei Shinohara
- Graduate School of Technology, Industrial and Social Science, Tokushima University, Minamijosanjimacho-2, Tokushima 770-8506, Japan
| | - Yuki Komiya
- Graduate School of Technology, Industrial and Social Science, Tokushima University, Minamijosanjimacho-2, Tokushima 770-8506, Japan
| | - Kashin Morimoto
- Graduate School of Technology, Industrial and Social Science, Tokushima University, Minamijosanjimacho-2, Tokushima 770-8506, Japan
| | - Yoshio Endo
- Central Research Resource Branch, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Minoru Terashima
- Division of Functional Geneomics, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Takeshi Suzuki
- Division of Functional Geneomics, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Takahisa Takino
- Division of Education for Global Standard, Institute of Liberal Arts and Science, Kanazawa University Kakuma-machi, Kanazawa 920-1192, Japan
| | - Itasu Ninomiya
- Director of Central Medical Center and Department of Surgery, Fukui Prefectural Hospital, Yotsui-2, Fukui 910-0846, Japan
| | - Hisatsugu Yamada
- Graduate School of Technology, Industrial and Social Science, Tokushima University, Minamijosanjimacho-2, Tokushima 770-8506, Japan
| | - Yoshihiro Uto
- Graduate School of Technology, Industrial and Social Science, Tokushima University, Minamijosanjimacho-2, Tokushima 770-8506, Japan.
| |
Collapse
|
8
|
Skandalis SS. CD44 Intracellular Domain: A Long Tale of a Short Tail. Cancers (Basel) 2023; 15:5041. [PMID: 37894408 PMCID: PMC10605500 DOI: 10.3390/cancers15205041] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
CD44 is a single-chain transmembrane receptor that exists in multiple forms due to alternative mRNA splicing and post-translational modifications. CD44 is the main cell surface receptor of hyaluronan as well as other extracellular matrix molecules, cytokines, and growth factors that play important roles in physiological processes (such as hematopoiesis and lymphocyte homing) and the progression of various diseases, the predominant one being cancer. Currently, CD44 is an established cancer stem cell marker in several tumors, implying a central functional role in tumor biology. The present review aims to highlight the contribution of the CD44 short cytoplasmic tail, which is devoid of any enzymatic activity, in the extraordinary functional diversity of the receptor. The interactions of CD44 with cytoskeletal proteins through specific structural motifs within its intracellular domain drives cytoskeleton rearrangements and affects the distribution of organelles and transport of molecules. Moreover, the CD44 intracellular domain specifically interacts with various cytoplasmic effectors regulating cell-trafficking machinery, signal transduction pathways, the transcriptome, and vital cell metabolic pathways. Understanding the cell type- and context-specificity of these interactions may unravel the high complexity of CD44 functions and lead to novel improved therapeutic interventions.
Collapse
Affiliation(s)
- Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| |
Collapse
|
9
|
Zheng S, An S, Luo Y, Vithran DTA, Yang S, Lu B, Deng Z, Li Y. HYBID in osteoarthritis: Potential target for disease progression. Biomed Pharmacother 2023; 165:115043. [PMID: 37364478 DOI: 10.1016/j.biopha.2023.115043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/07/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023] Open
Abstract
HYBID is a new hyaluronan-degrading enzyme and exists in various cells of the human body. Recently, HYBID was found to over-express in the osteoarthritic chondrocytes and fibroblast-like synoviocytes. According to these researches, high level of HYBID is significantly correlated with cartilage degeneration in joints and hyaluronic acid degradation in synovial fluid. In addition, HYBID can affect inflammatory cytokine secretion, cartilage and synovium fibrosis, synovial hyperplasia via multiple signaling pathways, thereby exacerbating osteoarthritis. Based on the existing research of HYBID in osteoarthritis, HYBID can break the metabolic balance of HA in joints through the degradation ability independent of HYALs/CD44 system and furthermore affect cartilage structure and mechanotransduction of chondrocytes. In particular, in addition to HYBID itself being able to trigger some signaling pathways, we believe that low-molecular-weight hyaluronan produced by excess degradation can also stimulate some disease-promoting signaling pathways by replacing high-molecular-weight hyaluronan in joints. The specific role of HYBID in osteoarthritis is gradually revealed, and the discovery of HYBID raises the new way to treat osteoarthritis. In this review, the expression and basic functions of HYBID in joints were summarized, and reveal potential role of HYBID as a key target in treatment for osteoarthritis.
Collapse
Affiliation(s)
- Shengyuan Zheng
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, China
| | - Senbo An
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yan Luo
- Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, China
| | - Djandan Tadum Arthur Vithran
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shaoqu Yang
- Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, China
| | - Bangbao Lu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Zhenhan Deng
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China.
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
10
|
Klement RJ, Sweeney RA. Metabolic factors associated with the prognosis of oligometastatic patients treated with stereotactic body radiotherapy. Cancer Metastasis Rev 2023; 42:927-940. [PMID: 37261610 DOI: 10.1007/s10555-023-10110-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/22/2023] [Indexed: 06/02/2023]
Abstract
Over the past two decades, it has been established that cancer patients with oligometastases, i.e., only a few detectable metastases confined to one or a few organs, may benefit from an aggressive local treatment approach such as the application of high-precision stereotactic body radiotherapy (SBRT). Specifically, some studies have indicated that achieving long-term local tumor control of oligometastases is associated with prolonged overall survival. This motivates investigations into which factors may modify the dose-response relationship of SBRT by making metastases more or less radioresistant. One such factor relates to the uptake of the positron emission tomography tracer 2-deoxy-2-[18F]fluoro-D-glucose (FDG) which reflects the extent of tumor cell glycolysis or the Warburg effect, respectively. Here we review the biological mechanisms how the Warburg effect drives tumor cell radioresistance and metastasis and draw connections to clinical studies reporting associations between high FDG uptake and worse clinical outcomes after SBRT for oligometastases. We further review the evidence for distinct metabolic phenotypes of metastases preferentially seeding to specific organs and their possible translation into distinct radioresistance. Finally, evidence that obesity and hyperglycemia also affect outcomes after SBRT will be presented. While delivered dose is the main determinant of a high local tumor control probability, there might be clinical scenarios when metabolic targeting could make the difference between achieving local control or not, for example when doses have to be compromised in order to spare neighboring high-risk organs, or when tumors are expected to be highly therapy-resistant due to heavy pretreatment such as chemotherapy and/or radiotherapy.
Collapse
Affiliation(s)
- Rainer J Klement
- Department of Radiotherapy and Radiation Oncology, Leopoldina Hospital Schweinfurt, Robert-Koch-Straße 10, 97422, Schweinfurt, Germany.
| | - Reinhart A Sweeney
- Department of Radiotherapy and Radiation Oncology, Leopoldina Hospital Schweinfurt, Robert-Koch-Straße 10, 97422, Schweinfurt, Germany
| |
Collapse
|
11
|
Yan T, Yang H, Xu C, Liu J, Meng Y, Jiang Q, Li J, Kang G, Zhou L, Xiao S, Xue Y, Xu J, Chen X, Che F. Inhibition of hyaluronic acid degradation pathway suppresses glioma progression by inducing apoptosis and cell cycle arrest. Cancer Cell Int 2023; 23:163. [PMID: 37568202 PMCID: PMC10422813 DOI: 10.1186/s12935-023-02998-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Abnormal hyaluronic acid (HA) metabolism is a major factor in tumor progression, and the metabolic regulation of HA mainly includes HA biosynthesis and catabolism. In glioma, abnormal HA biosynthesis is intimately involved in glioma malignant biological properties and the formation of immunosuppressive microenvironment; however, the role of abnormal HA catabolism in glioma remains unclear. METHODS HA catabolism is dependent on hyaluronidase. In TCGA and GEPIA databases, we found that among the 6 human hyaluronidases (HYAL1, HYAL2, HYAL3, HYAL4, HYALP1, SPAM1), only HYAL2 expression was highest in glioma. Next, TCGA and CGGA database were further used to explore the correlation of HYAL2 expression with glioma prognosis. Then, the mRNA expression and protein level of HYAL2 was determined by qRT-PCR, Western blot and Immunohistochemical staining in glioma cells and glioma tissues, respectively. The MTT, EdU and Colony formation assay were used to measure the effect of HYAL2 knockdown on glioma. The GSEA enrichment analysis was performed to explore the potential pathway regulated by HYAL2 in glioma, in addition, the HYAL2-regulated signaling pathways were detected by flow cytometry and Western blot. Finally, small molecule compounds targeting HYAL2 in glioma were screened by Cmap analysis. RESULTS In the present study, we confirmed that Hyaluronidase 2 (HYAL2) is abnormally overexpressed in glioma. Moreover, we found that HYAL2 overexpression is associated with multiple glioma clinical traits and acts as a key indicator for glioma prognosis. Targeting HYAL2 could inhibit glioma progression by inducing glioma cell apoptosis and cell cycle arrest. CONCLUSION Collectively, these observations suggest that HYAL2 overexpression could promote glioma progression. Thus, treatments that disrupt HA catabolism by altering HYAL2 expression may serve as effective strategies for glioma treatment.
Collapse
Affiliation(s)
- Tao Yan
- Central Laboratory, Linyi People's Hospital, Guangzhou University of Chinese Medicine, Linyi, 276000, Shandong Province, China
- Linyi Key Laboratory of Neurophysiology, Linyi People's Hospital, Linyi, 276000, Shandong Province, China
| | - He Yang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Caixia Xu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Junsi Liu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Yun Meng
- Central Laboratory, Linyi People's Hospital, Guangzhou University of Chinese Medicine, Linyi, 276000, Shandong Province, China
- Linyi Key Laboratory of Neurophysiology, Linyi People's Hospital, Linyi, 276000, Shandong Province, China
| | - Qing Jiang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Jinxing Li
- Department of Neurosurgery, Linyi People's Hospital, Linyi, 276000, Shandong Province, China
| | - Guiqiong Kang
- Central Laboratory, Linyi People's Hospital, Guangzhou University of Chinese Medicine, Linyi, 276000, Shandong Province, China
- Linyi Key Laboratory of Neurophysiology, Linyi People's Hospital, Linyi, 276000, Shandong Province, China
| | - Liangjian Zhou
- Scientific Research Management Office, Linyi People's Hospital, Linyi, 276000, Shandong Province, China
| | - Shuai Xiao
- Scientific Research Management Office, Linyi People's Hospital, Linyi, 276000, Shandong Province, China
| | - Yanpeng Xue
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Jiayi Xu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Xin Chen
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China.
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.
| | - Fengyuan Che
- Central Laboratory, Linyi People's Hospital, Guangzhou University of Chinese Medicine, Linyi, 276000, Shandong Province, China.
- Linyi Key Laboratory of Neurophysiology, Linyi People's Hospital, Linyi, 276000, Shandong Province, China.
- Department of Neurology, Linyi People's Hospital, Linyi, 276000, Shandong Province, China.
| |
Collapse
|
12
|
Kaur S, Balakrishnan B, Mallia MB, Keshari R, Hassan PA, Banerjee R. Technetium-99m labeled core shell hyaluronate nanoparticles as tumor responsive, metastatic skeletal lesion targeted combinatorial theranostics. Carbohydr Polym 2023; 312:120840. [PMID: 37059565 DOI: 10.1016/j.carbpol.2023.120840] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/15/2023] [Accepted: 03/20/2023] [Indexed: 04/07/2023]
Abstract
Achieving target specific delivery of chemotherapeutics in metastatic skeletal lesions remains a major challenge. Towards this, a dual drug loaded, radiolabeled multi-trigger responsive nanoparticles having partially oxidized hyaluronate (HADA) conjugated to alendronate shell and palmitic acid core were developed. While the hydrophobic drug, celecoxib was encapsulated in the palmitic acid core, the hydrophilic drug, doxorubicin hydrochloride was linked to the shell via a pH responsive imine linkage. Hydroxyapatite binding studies showed affinity of alendronate conjugated HADA nanoparticles to bones. Enhanced cellular uptake of the nanoparticles was achieved via HADA-CD44 receptor binding. HADA nanoparticles demonstrated trigger responsive release of encapsulated drugs in the presence of hyaluronidase, pH and glucose, present in excess in the tumor microenvironment. Efficacy of the nanoparticles for combination chemotherapy was established by >10-fold reduction in IC50 of drug loaded particles with a combination index of 0.453, as compared to free drugs in MDA-MB-231 cells. The nanoparticles could be radiolabeled with the gamma emitting radioisotope technetium-99m (99mTc) through a simple, 'chelator free', procedure with excellent radiochemical purity (RCP) (>90 %) and in vitro stability. 99mTc-labeled drug loaded nanoparticles reported herein constitutes a promising theranostic agent to target metastatic bone lesions. STATEMENT OF HYPOTHESES: Technetium-99m labeled, alendronate conjugated, dual targeting, tumor responsive, hyaluronate nanoparticle for tumor specific drug release and enhanced therapeutic effect, with real-time in vivo monitoring.
Collapse
Affiliation(s)
- Shahdeep Kaur
- Nanomedicine Laboratory, Department of Biosciences & Bioengineering, Indian Institute of Technology, Bombay, Mumbai 400076, India
| | - Biji Balakrishnan
- Nanomedicine Laboratory, Department of Biosciences & Bioengineering, Indian Institute of Technology, Bombay, Mumbai 400076, India; Nanotherapeutics & Biosensor Section, Chemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India.
| | - Madhava B Mallia
- Radiopharmaceutical Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Mumbai 400094, India
| | - Roshan Keshari
- Nanomedicine Laboratory, Department of Biosciences & Bioengineering, Indian Institute of Technology, Bombay, Mumbai 400076, India
| | - P A Hassan
- Nanotherapeutics & Biosensor Section, Chemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Mumbai 400094, India
| | - Rinti Banerjee
- Nanomedicine Laboratory, Department of Biosciences & Bioengineering, Indian Institute of Technology, Bombay, Mumbai 400076, India
| |
Collapse
|
13
|
Ahmad SMS, Nazar H, Rahman MM, Rusyniak RS, Ouhtit A. ITGB1BP1, a Novel Transcriptional Target of CD44-Downstream Signaling Promoting Cancer Cell Invasion. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:373-380. [PMID: 37252376 PMCID: PMC10225144 DOI: 10.2147/bctt.s404565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/25/2023] [Indexed: 05/31/2023]
Abstract
Breast cancer (BC) is the most common malignancy worldwide and has a poor prognosis, because it begins in the breast and disseminates to lymph nodes and distant organs. While invading, BC cells acquire aggressive characteristics from the tumor microenvironment through several mechanisms. Thus, understanding the mechanisms underlying the process of BC cell invasion can pave the way towards the development of targeted therapeutics focused on metastasis. We have previously reported that the activation of CD44 receptor with its major ligand hyaluronan (HA) promotes BC metastasis to the liver in vivo. Next, a gene expression profiling microarray analysis was conducted to identify and validate CD44-downstream transcriptional targets mediating its pro-metastatic function from RNA samples collected from Tet CD44-induced versus control MCF7-B5 cells. We have already validated a number of novel CD44-target genes and published their underlying signaling pathways in promoting BC cell invasion. From the same microarray analysis, Integrin subunit beta 1 binding protein 1 (ITGB1BP1) was also identified as a potential CD44-target gene that was upregulated (2-fold) upon HA activation of CD44. This report will review the lines of evidence collected from the literature to support our hypothesis, and further discuss the possible mechanisms linking HA activation of CD44 to its novel potential transcriptional target ITGB1BP1.
Collapse
Affiliation(s)
- Salma M S Ahmad
- Biological Sciences Program, Department of Biological and Environmental Sciences, College of Arts and Science, Qatar University, Doha, Qatar
| | - Hanan Nazar
- Biological Sciences Program, Department of Biological and Environmental Sciences, College of Arts and Science, Qatar University, Doha, Qatar
| | - Md Mizanur Rahman
- Biological Sciences Program, Department of Biological and Environmental Sciences, College of Arts and Science, Qatar University, Doha, Qatar
| | - Radoslaw Stefan Rusyniak
- Biological Sciences Program, Department of Biological and Environmental Sciences, College of Arts and Science, Qatar University, Doha, Qatar
| | - Allal Ouhtit
- Biological Sciences Program, Department of Biological and Environmental Sciences, College of Arts and Science, Qatar University, Doha, Qatar
| |
Collapse
|
14
|
Carvalho AM, Reis RL, Pashkuleva I. Hyaluronan Receptors as Mediators and Modulators of the Tumor Microenvironment. Adv Healthc Mater 2023; 12:e2202118. [PMID: 36373221 PMCID: PMC11469756 DOI: 10.1002/adhm.202202118] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/28/2022] [Indexed: 11/16/2022]
Abstract
The tumor microenvironment (TME) is a dynamic and complex matter shaped by heterogenous cancer and cancer-associated cells present at the tumor site. Hyaluronan (HA) is a major TME component that plays pro-tumorigenic and carcinogenic functions. These functions are mediated by different hyaladherins expressed by cancer and tumor-associated cells triggering downstream signaling pathways that determine cell fate and contribute to TME progression toward a carcinogenic state. Here, the interaction of HA is reviewed with several cell-surface hyaladherins-CD44, RHAMM, TLR2 and 4, LYVE-1, HARE, and layilin. The signaling pathways activated by these interactions and the respective response of different cell populations within the TME, and the modulation of the TME, are discussed. Potential cancer therapies via targeting these interactions are also briefly discussed.
Collapse
Affiliation(s)
- Ana M. Carvalho
- 3Bs Research Group, I3Bs ‐ Research Institute on Biomaterials Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineBarco4805‐017Portugal
- ICVS/3B's – PT Government Associate LaboratoryUniversity of MinhoBraga4710‐057Portugal
| | - Rui L. Reis
- 3Bs Research Group, I3Bs ‐ Research Institute on Biomaterials Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineBarco4805‐017Portugal
- ICVS/3B's – PT Government Associate LaboratoryUniversity of MinhoBraga4710‐057Portugal
| | - Iva Pashkuleva
- 3Bs Research Group, I3Bs ‐ Research Institute on Biomaterials Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineBarco4805‐017Portugal
- ICVS/3B's – PT Government Associate LaboratoryUniversity of MinhoBraga4710‐057Portugal
| |
Collapse
|
15
|
Drygalski K, Lecoutre S, Clément K, Dugail I. Hyaluronan in Adipose Tissue, Metabolic Inflammation, and Diabetes: Innocent Bystander or Guilty Party? Diabetes 2023; 72:159-169. [PMID: 36668999 DOI: 10.2337/db22-0676] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/03/2022] [Indexed: 01/21/2023]
Abstract
Hyaluronic acid, or hyaluronan (HA), is a nonsulfated glucosaminoglycan that has long been recognized for its hydrophilic properties and is widely used as a dermal filler. Despite much attention given to the study of other extracellular matrix (ECM) components, in the field of ECM properties and their contribution to tissue fibroinflammation, little is known of HA's potential role in the extracellular milieu. However, recent studies suggest that it is involved in inflammatory response, diet-induced insulin resistance, adipogenesis, and autoimmunity in type 1 diabetes. Based on its unique physical property as a regulator of osmotic pressure, we emphasize underestimated implications in adipose tissue function, adipogenesis, and obesity-related dysfunction.
Collapse
Affiliation(s)
- Krzysztof Drygalski
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
- Clinical Research Center, Medical University of Bialystok, Bialystok, Poland
| | - Simon Lecoutre
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| | - Karine Clément
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
- Nutrition Department, Assistance Publique Hôpitaux de Paris, Centre de Recherche en Nutrition Humaine Ile-de-France, Pitié-Salpêtrière Hospital, Paris, France
| | - Isabelle Dugail
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| |
Collapse
|
16
|
Shaban NZ, El-Faham AA, Abu-Serie MM, Habashy NH. Targeting apoptosis in MCF-7 and Ehrlich ascites carcinoma cells by saponifiable fractions from green and black Vitis vinifera seed oil. Biomed Pharmacother 2023; 157:114017. [PMID: 36395612 DOI: 10.1016/j.biopha.2022.114017] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 11/15/2022] Open
Abstract
Grape seed (GS) oil is one of the potential functional foods. For the first time, we evaluated the therapeutic effects of GS oil saponifiable (Sap)-fraction from black (BSap) and green (GSap) grapes on MCF-7 cells and Ehrlich ascites carcinoma (EAC) in mice. The fatty acid composition of BSap and GSap was determined using gas chromatography-mass spectrometry analysis. Approximately twelve distinct fatty acids were detected in BSap and eleven in GSap. BSap showed a greater cytotoxic effect on MCF-7 cells than GSap did by inducing apoptosis and reducing inflammation, while both grape fractions had superior potency to 5-FU. Furthermore, BSap massively boosted apoptosis and lowered redox potential (Eh) and CD44+ cells in EAC cells of EAC-bearing mice more than GSap, and both fractions were more efficient than 5-FU. Blood tests and liver histopathology revealed significant improvement in EAC-induced pathological alterations with these fractions. The in silico analysis implied the competitive inhibitory impacts of the most abundant fatty acid composites in BSap and GSap on cancer-metastasis-associated proteases (cathepsin B and MMP9). Also, this analysis predicted that the apoptotic action of these Sap fractions is independent of the 5'AMP-activated protein kinase. Therefore, grape Sap-fraction, especially BSap, may be a useful agent for cancer prevention.
Collapse
Affiliation(s)
- Nadia Z Shaban
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria 21511, Egypt
| | - Ashraf A El-Faham
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria 21511, Egypt
| | - Marwa M Abu-Serie
- Department of Medical Biotechnology, Genetic Engineering, and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg EL-Arab, Alexandria 21934, Egypt.
| | - Noha H Habashy
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria 21511, Egypt.
| |
Collapse
|
17
|
Zheng X, Wang B, Tang X, Mao B, Zhang Q, Zhang T, Zhao J, Cui S, Chen W. Absorption, metabolism, and functions of hyaluronic acid and its therapeutic prospects in combination with microorganisms: A review. Carbohydr Polym 2023; 299:120153. [PMID: 36876779 DOI: 10.1016/j.carbpol.2022.120153] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 09/21/2022] [Accepted: 09/21/2022] [Indexed: 11/25/2022]
Abstract
Hyaluronic acid (HA) is key to the stability of the internal environment of tissues. HA content in tissues gradually decreases with age, causing age-related health problems. Exogenous HA supplements are used to prevent or treat these problems including skin dryness and wrinkles, intestinal imbalance, xerophthalmia, and arthritis after absorption. Moreover, some probiotics are able to promote endogenous HA synthesis and alleviate symptoms caused by HA loss, thus introducing potential preventative or therapeutic applications of HA and probiotics. Here, we review the oral absorption, metabolism, and biological function of HA as well as the potential role of probiotics and HA in increasing the efficacy of HA supplements.
Collapse
Affiliation(s)
- Xueli Zheng
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Botao Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Bloomage Biotechnology Co., Ltd, Jinan 250000, China
| | - Xin Tang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Bingyong Mao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Qiuxiang Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Tianmeng Zhang
- Bloomage Biotechnology Co., Ltd, Jinan 250000, China; School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Shumao Cui
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
18
|
Beltran AS, King KE, La J, Reipolska A, Young KA. Short communication: Photoperiod impacts ovarian extracellular matrix and metabolic gene expression in Siberian hamsters. Comp Biochem Physiol A Mol Integr Physiol 2022; 274:111302. [PMID: 36041709 PMCID: PMC11285357 DOI: 10.1016/j.cbpa.2022.111302] [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: 04/02/2022] [Revised: 08/13/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022]
Abstract
Ovarian cyclicity is variable in adult Siberian hamsters (Phodopus sungorus), who respond to long breeding season photoperiods with follicle development and ovulation, while short photoperiods typical of the non-breeding season induce gonadal atrophy. Recent RNAseq results identified ovarian matrix components and regulators of metabolism as differentially regulated by photoperiod; however, the impact of photoperiod across a full cycle of ovarian regression and recrudescence had not been explored for additional regulators of ovarian metabolism and extracellular matrix components. We hypothesized that matrix and metabolism-related genes would be expressed differentially across photoperiods that mimic breeding and non-breeding season daylengths. Hamsters were housed in one of four photoperiod groups: long day (16 h of light per day: 8 h of dark; LD, controls), short day regressed (8 L:16D; SD, regressed), and females exposed to SD then transferred to LD to stimulate return of ovarian function for 2 (early recrudescence), or 8 (late recrudescence) weeks. Plasma leptin concentrations along with expression of ovarian versican and liver-receptor homolog-1/Nr582 mRNA decreased in SD compared to LD and late recrudescence, while vimentin mRNA expression peaked in early and late recrudescence. Ovarian expression of fibronectin and extracellular matrix protein-1 was low in LD ovaries and increased in regressed and recrudescing groups. Expression of hyaluronidase-2, nectin-2, liver-X receptors-α and-β, and adiponectin mRNA peaked in late recrudescence, with no changes noted for adiponectin receptor-1 and -2. The results offer a first look at the parallels between expression of these genes and the dynamic remodeling that occurs during ovarian regression and recrudescence.
Collapse
Affiliation(s)
- Arianna S Beltran
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, United States of America
| | - Kristen E King
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, United States of America
| | - Josephine La
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, United States of America
| | - Anastasiia Reipolska
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, United States of America
| | - Kelly A Young
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, United States of America.
| |
Collapse
|
19
|
Lin J, Liu G, Chen L, Kwok HF, Lin Y. Targeting lactate-related cell cycle activities for cancer therapy. Semin Cancer Biol 2022; 86:1231-1243. [PMID: 36328311 DOI: 10.1016/j.semcancer.2022.10.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
Abstract
Lactate has long been considered as a metabolic by-product of aerobic glycolysis for cancer. However, more and more studies have shown that lactate can regulate cancer progression via multiple mechanisms such as cell cycle regulation, immune suppression, energy metabolism and so on. A recent discovery of lactylation attracted a lot of attention and is already a hot topic in the cancer field. In this review, we summarized the latest functions of lactate and its underlying mechanisms in cancer. We also included our analysis of protein lactylation in different rat organs and compared them with other published lactylation data. The unresolved challenges in this field were discussed, and the potential application of these new discoveries of lactate-related cell cycle activities for cancer target therapy was speculated.
Collapse
Affiliation(s)
- Jia Lin
- Central Laboratory at the Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Collaborative Innovation Center for Rehabilitation Technology, the Institute of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, China; Collaborative Innovation Center for Rehabilitation Technology, the Institute of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, China
| | - Geng Liu
- Laboratory of Cancer Biology, Department of Oncology, University of Oxford, UK
| | - Lidian Chen
- Collaborative Innovation Center for Rehabilitation Technology, the Institute of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, China.
| | - Hang Fai Kwok
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR; MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau SAR; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR.
| | - Yao Lin
- Central Laboratory at the Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Collaborative Innovation Center for Rehabilitation Technology, the Institute of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, China; Fujian-Macao Science and Technology Cooperation Base of Traditional Chinese Medicine-Oriented Chronic Disease Prevention and Treatment, Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, China.
| |
Collapse
|
20
|
Zhang Y, Peng Q, Zheng J, Yang Y, Zhang X, Ma A, Qin Y, Qin Z, Zheng X. The function and mechanism of lactate and lactylation in tumor metabolism and microenvironment. Genes Dis 2022. [PMID: 37492749 PMCID: PMC10363641 DOI: 10.1016/j.gendis.2022.10.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Lactate is an end product of glycolysis. Owing to the lactate shuttle concept introduced in the early 1980s, increasing researchers indicate lactate as a critical energy source for mitochondrial respiration and as a precursor of gluconeogenesis. Lactate also acts as a multifunctional signaling molecule through receptors expressed in various cells, resulting in diverse biological consequences including decreased lipolysis, immune regulation, and anti-inflammation wound healing, and enhanced exercise performance in association with the gut microbiome. Furthermore, increasing evidence reveals that lactate contributes to epigenetic gene regulation by lactylating lysine residues of histones, which accounts for its key role in immune modulation and maintenance of homeostasis. Here, we summarize the function and mechanism of lactate and lactylation in tumor metabolism and microenvironment.
Collapse
|
21
|
Zhao M, Liu J, Tang Y, Zhang L, Ge X, Chen M, Wen Q, Zhu L, Ma Q. Hyaluronidase responsive second near-infrared fluorescent nanocomplex for combined HER2 blockade and chemotherapy of HER2+ breast cancer. BIOMATERIALS ADVANCES 2022; 141:213115. [PMID: 36115156 DOI: 10.1016/j.bioadv.2022.213115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/22/2022] [Accepted: 09/07/2022] [Indexed: 06/15/2023]
Abstract
The human epidermal growth factor receptor-2-positive (HER2+) type is aggressive and has poor prognosis. Although anti-HER2 therapy alone or in combination with other treatment regimens showed significant improvement in survival outcomes, breast cancer patients are still suffering from tumor relapse and severe dose-limiting side effects. Thus, there is still an unmet challenge to develop effective therapeutic agents for HER2+ breast cancer treatment with minimized side effects. Herein, we produced a stimuli-responsive and tumor-targeted hyaluronic acid (HA) nanocomplex that combined HER2 blockade and chemotherapy for effective HER2+ breast cancer therapy. A hydrophobic NIR-II dye, IR1048, was covalently linked with HA to form a spherical HA-IR1048 nanoparticle (HINP), with Herceptin conjugated on the surface and paclitaxel (PTX) encapsulated inside. The fluorescent signals from the yielding Her-HINP/PTX are quenched originally, but a strong NIR-II signal is generated when HINP is degraded by the hyaluronidase that is overexpressed in breast tumors, thus allowing the tracking and visualization of Herceptin and PTX accumulation. Her-HINP/PTX peaked in HER2+ tumors at 24 h post injection as imaged by NIR-II fluorescent imaging. A significantly improved tumor growth inhibition effect was observed after five systemic treatments compared to single PTX (3.71 ± 0.41 times) or Herceptin (5.98 ± 0.51 times) treatment in a HER2-overexpressed breast cancer mouse model with prolonged survival. Collectively, the designed Her-HINP/PTX presents a new hyaluronidase-responsive and HER2 blockade nanoformulation that can visualize the accumulation of nanocomplexes and release drugs inside tumors for combined HER2+ breast cancer therapy with a great promise for translational study. STATEMENT OF SIGNIFICANCE: The high expressions of a protein called human epidermal growth factor receptor 2 (HER2) in breast tumors make this subtype of cancer aggressive. Currently, chemotherapy combined with a HER2 antibody, Herceptin, is a preferred approach for HER2-positive breast cancer therapy. However, these breast cancer patients still suffer from tumor relapse and severe side effects because various therapeutic agents have inherent different biodistributions, resulting in insufficient treatment effects and unfavorable normal organ uptake of these therapeutic agents. Herein, we produced a nanocomplex carrying both Herceptin and chemotherapy drug to simultaneously deliver two drugs into tumors for efficient HER2+ tumor treatment with minimized side effects, providing new insights for designing a combined therapy strategy.
Collapse
Affiliation(s)
- Min Zhao
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, China
| | - Junzhi Liu
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, China
| | - Yuting Tang
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, China
| | - Lumeng Zhang
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, China
| | - Xiaoguang Ge
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, China; MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, China
| | - Minglong Chen
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, China
| | - Qiang Wen
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, China.
| | - Lei Zhu
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, China.
| | - Qingjie Ma
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, China.
| |
Collapse
|
22
|
MacLeod R, Chan FV, Yuan H, Ye X, Sin YJA, Vitelli TM, Cucu T, Leung A, Baljak I, Osinski S, Fu Y, Jung GID, Amar A, DeAngelis PL, Hellman U, Cowman MK. Selective isolation of hyaluronan by solid phase adsorption to silica. Anal Biochem 2022; 652:114769. [PMID: 35660507 PMCID: PMC9589902 DOI: 10.1016/j.ab.2022.114769] [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: 12/05/2021] [Revised: 04/28/2022] [Accepted: 05/27/2022] [Indexed: 11/01/2022]
Abstract
A solid phase adsorption method for selective isolation of hyaluronan (HA) from biological samples is presented. Following enzymatic degradation of protein, HA can be separated from sulfated glycosaminoglycans, other unsulfated glycosaminoglycans, nucleic acids, and proteolytic fragments by adsorption to amorphous silica at specific salt concentrations. The adsorbed HA can be released from silica using neutral and basic aqueous solutions. HA ranging in size from ∼9 kDa to MDa polymers has been purified by this method from human serum and conditioned medium of cultured cells.
Collapse
Affiliation(s)
- Rebecca MacLeod
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, 433 First Avenue, 9thfloor, New York, NY, 10010, USA.
| | - Fok Vun Chan
- Echelon Biosciences Inc., 675 Arapeen Drive, Suite 302, Salt Lake City, UT, 84108, USA.
| | - Han Yuan
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, 6 Metrotech Center, Brooklyn, NY, 11201, USA.
| | - Xin Ye
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, 6 Metrotech Center, Brooklyn, NY, 11201, USA.
| | - Yun Jin Ashley Sin
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, 433 First Avenue, 9thfloor, New York, NY, 10010, USA.
| | - Teraesa M Vitelli
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, 433 First Avenue, 9thfloor, New York, NY, 10010, USA.
| | - Tudor Cucu
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, 6 Metrotech Center, Brooklyn, NY, 11201, USA.
| | - Annie Leung
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, 6 Metrotech Center, Brooklyn, NY, 11201, USA.
| | - Irene Baljak
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, 433 First Avenue, 9thfloor, New York, NY, 10010, USA.
| | - Samantha Osinski
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, 6 Metrotech Center, Brooklyn, NY, 11201, USA.
| | - Yuhong Fu
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, 6 Metrotech Center, Brooklyn, NY, 11201, USA.
| | - Gyu Ik Daniel Jung
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, 6 Metrotech Center, Brooklyn, NY, 11201, USA.
| | - Anant Amar
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, 6 Metrotech Center, Brooklyn, NY, 11201, USA.
| | - Paul L DeAngelis
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd., Oklahoma, OK, 73104, USA.
| | - Urban Hellman
- Department of Public Health and Clinical Medicine, Umeå University, SE-901 87, Umeå, Sweden.
| | - Mary K Cowman
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, 433 First Avenue, 9thfloor, New York, NY, 10010, USA; Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, 6 Metrotech Center, Brooklyn, NY, 11201, USA.
| |
Collapse
|
23
|
Parnigoni A, Viola M, Karousou E, Rovera S, Giaroni C, Passi A, Vigetti D. ROLE OF HYALURONAN IN PATHOPHYSIOLOGY OF VASCULAR1 ENDOTHELIAL AND SMOOTH MUSCLE CELLS. Am J Physiol Cell Physiol 2022; 323:C505-C519. [PMID: 35759431 DOI: 10.1152/ajpcell.00061.2022] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
One of the main components of the extracellular matrix (ECM) of the blood vessel is hyaluronic acid or hyaluronan (HA). It is a ubiquitous polysaccharide belonging to the family of glycosaminoglycans, but, differently from other proteoglycan-associated glycosaminoglycans, it is synthesized on the plasma membrane by a family of three HA synthases (HAS). HA can be released as a free polymer in the extracellular space or remain associated with the membrane in the pericellular space via HAS or via binding proteins. In fact, several cell surface proteins can interact with HA working as HA receptors like CD44, RHAMM, and LYVE-1. In physiological conditions, HA is localized in the glycocalyx and in the adventitia and is responsible for the loose and hydrated vascular structure favoring flexibility and allowing the stretching of vessels in response to mechanical forces. During atherogenesis, ECM undergoes dramatic alterations which have a crucial role in lipoprotein retention and in triggering multiple signaling cascades that wake up cells from their quiescent status. HA becomes highly present in the media and neointima favoring smooth muscle cells dedifferentiation, migration, and proliferation that strongly contribute to vessel wall thickening. Further, HA is able to modulate immune cell recruitment both within the vessel wall and on the endothelial cell layer. This review is focused on the effects of HA on vascular cell behavior.
Collapse
Affiliation(s)
- Arianna Parnigoni
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Manuela Viola
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Evgenia Karousou
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Simona Rovera
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Cristina Giaroni
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Davide Vigetti
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| |
Collapse
|
24
|
Karalis T, Skandalis SS. Hyaluronan network: a driving force in cancer progression. Am J Physiol Cell Physiol 2022; 323:C145-C158. [PMID: 35649255 DOI: 10.1152/ajpcell.00139.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hyaluronan is one of the most abundant macromolecules of the extracellular matrix and regulates several physiological cell and tissue properties. However, hyaluronan has been shown to accumulate together with its receptors in various cancers. In tumors, accumulation of hyaluronan system components (hyaluronan synthesizing/degrading enzymes and interacting proteins) associates with poor outcomes of the patients. In this article, we review the main roles of hyaluronan in normal physiology and cancer, and further discuss the targeting of hyaluronan system as an applicable therapeutic strategy.
Collapse
Affiliation(s)
- Theodoros Karalis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Spyros S Skandalis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| |
Collapse
|
25
|
Kciuk M, Gielecińska A, Mujwar S, Mojzych M, Marciniak B, Drozda R, Kontek R. Targeting carbonic anhydrase IX and XII isoforms with small molecule inhibitors and monoclonal antibodies. J Enzyme Inhib Med Chem 2022; 37:1278-1298. [PMID: 35506234 PMCID: PMC9090362 DOI: 10.1080/14756366.2022.2052868] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Carbonic anhydrases IX and CAXII (CAIX/CAXII) are transmembrane zinc metalloproteins that catalyze a very basic but crucial physiological reaction: the conversion of carbon dioxide into bicarbonate with a release of the proton. CA, especially CAIX and CAXII isoforms gained the attention of many researchers interested in anticancer drug design due to pivotal functions of enzymes in the cancer cell metastasis and response to hypoxia, and their expression restricted to malignant cells. This offers an opportunity to develop new targeted therapies with fewer side effects. Continuous efforts led to the discovery of a series of diverse compounds with the most abundant sulphonamide derivatives. Here we review current knowledge considering small molecule and antibody-based targeting of CAIX/CAXII in cancer.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland.,Doctoral School of Exact and Natural Sciences, University of Lodz, Lodz, Poland
| | - Adrianna Gielecińska
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland
| | - Somdutt Mujwar
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Mariusz Mojzych
- Department of Chemistry, Siedlce University of Natural Sciences and Humanities, Siedlce, Poland
| | - Beata Marciniak
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland
| | - Rafał Drozda
- Department of Gastrointestinal Endoscopy, Wl. Bieganski Hospital, Lodz, Poland
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland
| |
Collapse
|
26
|
Lung Hyaluronasome: Involvement of Low Molecular Weight Ha (Lmw-Ha) in Innate Immunity. Biomolecules 2022; 12:biom12050658. [PMID: 35625586 PMCID: PMC9138743 DOI: 10.3390/biom12050658] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 02/01/2023] Open
Abstract
Hyaluronic acid (HA) is a major component of the extracellular matrix. It is synthesized by hyaluronan synthases (HAS) into high-molecular-weight chains (HMW-HA) that exhibit anti-inflammatory and immunomodulatory functions. In damaged, infected, and/or inflamed tissues, HMW-HA are degraded by hyaluronidases (HYAL) or reactive oxygen species (ROS) to give rise to low-molecular-weight HAs (LMW-HAs) that are potent pro-inflammatory molecules. Therefore, the size of HA regulates the balance of anti- or pro-inflammatory functions. The activities of HA depend also on its interactions with hyaladherins. HA synthesis, degradation, and activities through HA/receptors interactions define the hyaluronasome. In this review, a short overview of the role of high and low-molecular-weight HA polymers in the lungs is provided. The involvement of LMW-HA in pulmonary innate immunity via the activation of neutrophils, macrophages, dendritic cells, and epithelial cells is described to highlight LMW-HA as a therapeutic target in inflammatory respiratory diseases. Finally, the possibilities to counter LMW-HA’s deleterious effects in the lungs are discussed.
Collapse
|
27
|
Deguchi H, Yamashita T, Hiramoto N, Otsuki Y, Mukai A, Ueno M, Sotozono C, Kinoshita S, Hamuro J. Intracellular pH affects mitochondrial homeostasis in cultured human corneal endothelial cells prepared for cell injection therapy. Sci Rep 2022; 12:6263. [PMID: 35428816 PMCID: PMC9012833 DOI: 10.1038/s41598-022-10176-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 03/30/2022] [Indexed: 12/11/2022] Open
Abstract
This study aimed to uncover the mechanism responsible for the clinical efficacy of cell injection therapy with fully differentiated cultured cells. Analysis of polarized expression of ion transporters on cultured human corneal endothelial cells (CECs) subpopulations (SPs) was performed. The intracellular pH (pHi) between two CEC SPs, distinct in the proportion of differentiated cells, was measured, and the association with mitochondrial respiration homeostasis was investigated. The effects of the ion transporter inhibition by their selective inhibitors or siRNA transfection were also explored. Na+/K+-ATPase, Aquaporin 1, SLC4A11, NBCe1, NHE1 as transporters, and ZO-1, were all selectively expressed in differentiated SPs, but were almost null in the cell-state-transitioned SPs. We also confirmed that the pHi of CEC SPs affected their mitochondrial respiration by modulating the expression of these ion transporters via inhibitors or siRNA transfection. Ion and water transporters might participate in the maintenance of pHi and mitochondria homeostasis in differentiated SPs, which may contribute, combined with integral barrier functions, to efficient water efflux. The differences in intracellular pH between the two SPs is attributed to variations in the expression profile of specific ion transporters and mitochondrial functions, which may associate with the efficacy of the SPs in cell injection therapy.
Collapse
Affiliation(s)
- Hideto Deguchi
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan
| | - Tomoko Yamashita
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan
| | - Nao Hiramoto
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan
| | - Yohei Otsuki
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan
| | - Atsushi Mukai
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan
| | - Morio Ueno
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan
| | - Chie Sotozono
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan
| | - Shigeru Kinoshita
- Department of Frontier Medical Science and Technology for Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Junji Hamuro
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan.
| |
Collapse
|
28
|
Rivas F, Erxleben D, Smith I, Rahbar E, DeAngelis PL, Cowman MK, Hall AR. Methods for isolating and analyzing physiological hyaluronan: a review. Am J Physiol Cell Physiol 2022; 322:C674-C687. [PMID: 35196167 PMCID: PMC8977137 DOI: 10.1152/ajpcell.00019.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/22/2022] [Accepted: 02/22/2022] [Indexed: 01/01/2023]
Abstract
The carbohydrate hyaluronan (or hyaluronic acid, HA) is found in all human tissues and biofluids where it has wide-ranging functions in health and disease that are dictated by both its abundance and size. Consequently, hyaluronan evaluation in physiological samples has significant translational potential. Although the analytical tools and techniques for probing other biomolecules such as proteins and nucleic acids have become standard approaches in biochemistry, those available for investigating hyaluronan are less well established. In this review, we survey methods related to the assessment of native hyaluronan in biological specimens, including protocols for separating it from biological matrices and technologies for determining its concentration and molecular weight.
Collapse
Affiliation(s)
- Felipe Rivas
- Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Dorothea Erxleben
- Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Ian Smith
- Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Elaheh Rahbar
- Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Paul L DeAngelis
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Mary K Cowman
- Department of Biomedical Engineering, New York University Tandon School of Engineering, New York, New York
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, New York
| | - Adam R Hall
- Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
29
|
The Degradation of Hyaluronan in the Skin. Biomolecules 2022; 12:biom12020251. [PMID: 35204753 PMCID: PMC8961566 DOI: 10.3390/biom12020251] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/28/2022] [Accepted: 01/31/2022] [Indexed: 02/04/2023] Open
Abstract
Hyaluronan (HA) comprises a fundamental component of the extracellular matrix and participates in a variety of biological processes. Half of the total amount of HA in the human body is present in the skin. HA exhibits a dynamic turnover; its half-life in the skin is less than one day. Nevertheless, the specific participants in the catabolism of HA in the skin have not yet been described in detail, despite the essential role of HA in cutaneous biology. A deeper knowledge of the processes involved will act to support the development of HA-based topical and implantable materials and enhance the understanding of the various related pathological cutaneous conditions. This study aimed to characterize the distribution and activity of hyaluronidases and the other proteins involved in the degradation of HA in healthy human full-thickness skin, the epidermis and the dermis. Hyaluronidase activity was detected for the first time in healthy human skin. The degradation of HA occurred in lysates at an acidic pH. HA gel zymography revealed a single band corresponding to approximately 50 kDa. This study provided the first comprehensive view of the distribution of canonic HA-degrading proteins (HYAL1 and HYAL2) in human skin employing IHF and IHC. Furthermore, contrary to previous assumptions TMEM2, a novel hyaluronidase, as well as CEMIP, a protein involved in HA degradation, were localized in the human epidermis, as well as in the dermis.
Collapse
|
30
|
Takatani-Nakase T, Matsui C, Hosotani M, Omura M, Takahashi K, Nakase I. Hypoxia enhances motility and EMT through the Na +/H + exchanger NHE-1 in MDA-MB-231 breast cancer cells. Exp Cell Res 2022; 412:113006. [PMID: 34979106 DOI: 10.1016/j.yexcr.2021.113006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 12/23/2021] [Accepted: 12/30/2021] [Indexed: 12/15/2022]
Abstract
Breast cancer metastasis is the leading cause of cancer-related deaths. Hypoxia in the tumor mass is believed to trigger cell migration, which is involved in a crucial process of breast cancer metastasis. However, the molecular mechanisms underlying aggressive behavior under hypoxic conditions have not been fully elucidated. Here, we demonstrate the significant motility of MDA-MB-231 cells cultured under hypoxic conditions compared to that of cells cultured under normoxic conditions. MDA-MB-231 cells under hypoxic conditions showed a significant increase in Na+/H+ exchanger isoform 1 (NHE1) expression level, which was observed to co-locate in lamellipodia formation. Inhibition of NHE1 significantly suppressed the intracellular pH and the expression of mesenchymal markers, thereby blocking the high migration activity in hypoxia. Moreover, treatment with ciglitazone, a potent and selective peroxisome proliferator-activated receptor γ (PPARγ) agonist, modulated hypoxia-enhanced motion in cells via the repression of NHE1. These findings highlight that NHE1 is required for migratory activity through the enhancement of epithelial-mesenchymal transition (EMT) in MDA-MB-231 cells under hypoxic conditions, and we propose new drug repurposing strategies targeting hypoxia based on NHE1 suppression by effective usage of PPARγ agonists.
Collapse
Affiliation(s)
- Tomoka Takatani-Nakase
- Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, 11-68, Koshien Kyuban-cho, Nishinomiya, Hyogo, 663-8179, Japan; Institute for Bioscience, Mukogawa Women's University, 11-68, Koshien Kyuban-cho, Nishinomiya, Hyogo, 663-8179, Japan.
| | - Chihiro Matsui
- Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, 11-68, Koshien Kyuban-cho, Nishinomiya, Hyogo, 663-8179, Japan
| | - Maiko Hosotani
- Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, 11-68, Koshien Kyuban-cho, Nishinomiya, Hyogo, 663-8179, Japan
| | - Mika Omura
- Graduate School of Science, Osaka Prefecture University, 1-1, Gakuen-cho, Naka-ku, Sakai, Osaka, 599-8531, Japan
| | - Koichi Takahashi
- Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, 11-68, Koshien Kyuban-cho, Nishinomiya, Hyogo, 663-8179, Japan
| | - Ikuhiko Nakase
- Graduate School of Science, Osaka Prefecture University, 1-1, Gakuen-cho, Naka-ku, Sakai, Osaka, 599-8531, Japan; NanoSquare Research Institute, Osaka Prefecture University, 1-2, Gakuen-cho, Naka-ku, Sakai, Osaka, 599-8570, Japan.
| |
Collapse
|
31
|
Zakusilo FT, Kerry O'Banion M, Gelbard HA, Seluanov A, Gorbunova V. Matters of size: Roles of hyaluronan in CNS aging and disease. Ageing Res Rev 2021; 72:101485. [PMID: 34634492 PMCID: PMC8903057 DOI: 10.1016/j.arr.2021.101485] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 12/29/2022]
Abstract
Involvement of extracellular matrix (ECM) components in aging and age-related neurodegeneration is not well understood. The role of hyaluronan (HA), a major extracellular matrix glycosaminoglycan, in malignancy and inflammation is gaining new understanding. In particular, the differential biological effects of high molecular weight (HMW-HA) and low molecular weight hyaluronan (LMW-HA), and the mechanism behind such differences are being uncovered. Tightly regulated in the brain, HA can have diverse effects on cellular development, growth and degeneration. In this review, we summarize the homeostasis and signaling of HA in healthy tissue, discuss its distribution and ontogeny in the central nervous system (CNS), summarize evidence for its involvement in age-related neurodegeneration and Alzheimer Disease (AD), and assess the potential of HA as a therapeutic target in the CNS.
Collapse
Affiliation(s)
- Frances Tolibzoda Zakusilo
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA; Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - M Kerry O'Banion
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA; Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| | - Harris A Gelbard
- Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA; Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, NY, USA
| | - Andrei Seluanov
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| |
Collapse
|
32
|
Yamamoto T, Suzuki S, Fujii T, Mima Y, Watanabe K, Matsumoto M, Nakamura M, Fujita N. Efficacy of hyaluronic acid on intervertebral disc inflammation: An in vitro study using notochordal cell lines and human disc cells. J Orthop Res 2021; 39:2197-2208. [PMID: 33251629 DOI: 10.1002/jor.24933] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 11/09/2020] [Accepted: 11/25/2020] [Indexed: 02/04/2023]
Abstract
Hyaluronic acid (HA) is widely recognized as a therapeutic target and currently used in medicine. However, HA metabolism during intervertebral disc degeneration (IVDD) has not been completely elucidated. This study aimed to evaluate the efficacy of HA on intervertebral disc (IVD) inflammation and identify the main molecules modulating HA degradation in IVDs. To assess HA function in IVD cells in vitro, we treated human disc cells and U-CH1-N cells, a notochordal nucleus pulposus cell line, with HA or hyaluronidase. Real-time reverse transcription polymerase chain reaction (RT-PCR) and Western blot analysis showed that tumor necrosis factor alpha (TNF-α)-mediated induction of the expression of TNF-α and cyclooxygenase-2 (COX2) was clearly neutralized by HA treatment, and the expression of TNF-α and COX2 was significantly induced by hyaluronidase treatment in both cell types. Additionally, Western blot analysis showed that hyaluronidase-induced phosphorylation of p38 and Erk1/2, and that TNF-α-mediated phosphorylation of p38 and Erk1/2 was clearly reduced by HA addition. In degenerating human IVD samples, immunohistochemistry for hyaluronidase showed that the expression of hyaluronidases including HYAL1, HYAL2, and cell migration-inducing protein (CEMIP) tended to increase in accordance with IVDD. In particular, HYAL1 showed statistically significant differences. In vitro study also confirmed a similar phenomenon that TNF-α treatment increased both messenger RNA and protein expression in both cell types. Our results demonstrated that HA could potentially suppress IVDD by regulating p38 and Erk1/2 pathways, and that the expression of HYAL1 was correlated with IVDD progression. These findings indicated that HYAL1 would be a potential molecular target for suppressing IVDD by controlling HA metabolism.
Collapse
Affiliation(s)
- Tatsuya Yamamoto
- Department of Orthopaedic Surgery, School of Medicine, Keio University, Tokyo, Japan
| | - Satoshi Suzuki
- Department of Orthopaedic Surgery, School of Medicine, Keio University, Tokyo, Japan
| | - Takeshi Fujii
- Department of Orthopaedic Surgery, School of Medicine, Keio University, Tokyo, Japan.,Department of Orthopaedic Surgery, Saiseikai Yokohamashi Tobu Hospital, Kanagawa, Japan
| | - Yuichiro Mima
- Department of Orthopaedic Surgery, School of Medicine, Keio University, Tokyo, Japan.,Department of Orthopaedic Surgery, Kawasaki Municipal Hospital, Kanagawa, Japan
| | - Kota Watanabe
- Department of Orthopaedic Surgery, School of Medicine, Keio University, Tokyo, Japan
| | - Morio Matsumoto
- Department of Orthopaedic Surgery, School of Medicine, Keio University, Tokyo, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, School of Medicine, Keio University, Tokyo, Japan
| | - Nobuyuki Fujita
- Department of Orthopaedic Surgery, School of Medicine, Keio University, Tokyo, Japan.,Department of Orthopaedic Surgery, Fujita Health University, Toyoake, Aichi, Japan
| |
Collapse
|
33
|
Bychkov ML, Kirichenko AV, Shulepko MA, Mikhaylova IN, Kirpichnikov MP, Lyukmanova EN. Mambalgin-2 Inhibits Growth, Migration, and Invasion of Metastatic Melanoma Cells by Targeting the Channels Containing an ASIC1a Subunit Whose Up-Regulation Correlates with Poor Survival Prognosis. Biomedicines 2021; 9:1324. [PMID: 34680442 PMCID: PMC8533404 DOI: 10.3390/biomedicines9101324] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 02/04/2023] Open
Abstract
Melanoma is an aggressive cancer characterized by the acidification of the extracellular environment. Here, we showed for the first time that extracellular media acidification increases proliferation, migration, and invasion of patient-derived metastatic melanoma cells and up-regulates cell-surface expression of acid-sensitive channels containing the ASIC1a, α-ENaC, and γ-ENaC subunits. No influence of media acidification on these processes was found in normal keratinocytes. To control metastatic melanoma progression associated with the ASIC1a up-regulation, we proposed the ASIC1a inhibitor, -mambalgin-2 from Dendpoaspis polylepis venom. Recombinant analog of mambalgin-2 cancelled acidification-induced proliferation, migration, and invasion of metastatic melanoma cells, promoted apoptosis, and down-regulated cell-surface expression of prooncogenic factors CD44 and Frizzled 4 and phosphorylation of transcription factor SNAI. Confocal microscopy and affinity purification revealed that mambalgin-2 interacts with heterotrimeric ASIC1a/α-ENaC/γ-ENaC channels on the surface of metastatic melanoma cells. Using the mutant variant of mambalgin-2 with reduced activity toward ASIC1a, we confirmed that the principal molecular target of mambalgin-2 in melanoma cells is the ASIC1a subunit. Bioinformatic analysis confirmed up-regulation of the ASIC1 expression as a marker of poor survival prognosis for patients with metastatic melanoma. Thus, targeting ASIC1a by drugs such as mambalgin-2 could be a promising strategy for metastatic melanoma treatment.
Collapse
Affiliation(s)
- Maxim L. Bychkov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia; (M.L.B.); (A.V.K.); (M.A.S.); (M.P.K.)
| | - Artem V. Kirichenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia; (M.L.B.); (A.V.K.); (M.A.S.); (M.P.K.)
- Moscow Institute of Physics and Technology (State University), 141701 Dolgoprudny, Russia
| | - Mikhail A. Shulepko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia; (M.L.B.); (A.V.K.); (M.A.S.); (M.P.K.)
| | - Irina N. Mikhaylova
- Federal State Budgetary Institution “N.N. Blokhin National Medical Research Center of Oncology”, Ministry of Health of Russia, 115548 Moscow, Russia;
| | - Mikhail P. Kirpichnikov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia; (M.L.B.); (A.V.K.); (M.A.S.); (M.P.K.)
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Ekaterina N. Lyukmanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia; (M.L.B.); (A.V.K.); (M.A.S.); (M.P.K.)
- Moscow Institute of Physics and Technology (State University), 141701 Dolgoprudny, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
34
|
Garantziotis S. Modulation of hyaluronan signaling as a therapeutic target in human disease. Pharmacol Ther 2021; 232:107993. [PMID: 34587477 DOI: 10.1016/j.pharmthera.2021.107993] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/14/2022]
Abstract
The extracellular matrix is an active participant, modulator and mediator of the cell, tissue, organ and organismal response to injury. Recent research has highlighted the role of hyaluronan, an abundant glycosaminoglycan constituent of the extracellular matrix, in many fundamental biological processes underpinning homeostasis and disease development. From this basis, emerging studies have demonstrated the therapeutic potential of strategies which target hyaluronan synthesis, biology and signaling, with significant promise as therapeutics for a variety of inflammatory and immune diseases. This review summarizes the state of the art in this field and discusses challenges and opportunities in what could emerge as a new class of therapeutic agents, that we term "matrix biologics".
Collapse
Affiliation(s)
- Stavros Garantziotis
- Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA.
| |
Collapse
|
35
|
Wu ZL, Ertelt JM. Assays for hyaluronidases and heparanase using nonreducing end fluorophore-labeled hyaluronan and heparan sulfate proteoglycan. Glycobiology 2021; 31:1435-1443. [PMID: 34280262 DOI: 10.1093/glycob/cwab061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 01/30/2023] Open
Abstract
Glycosaminoglycans (GAGs), such as hyaluronan (HA) and heparan sulfate (HS), are a large group of polysaccharides found in the extracellular matrix and on the cell surface. The turnover of these molecules is controlled by de novo synthesis and catabolism through specific endoglycosidases, which are the keys to our understanding of the homeostasis of GAGs and could hold opportunities for therapeutic intervention. Herein, we describe assays for endoglycosidases using nonreducing end fluorophore-labeled GAGs, in which GAGs were labeled via incorporation of GlcNAz by specific synthases and cycloaddition of alkyne fluorophores and then digested with corresponding endoglycosidases. Assays of various HA-specific hyaluronidases (HYALs), including PH-20 or SPAM1, and HS-specific heparanase (HPSE) are presented. We demonstrated the distinctive pH profiles, substrate specificities and specific activities of these enzymes and provided evidence that both HYAL3 and HYAL4 are authentic hyaluronidases. In addition, while all HYALs are active on high-molecular-weight HA, they are active on low-molecular-weight HA. Subsequently, we defined a new way of measuring the activities of HYALs. Our results indicate that the activities of HYALs must be under strict pH regulation. Our quantitative methods of measuring the activity GAG endoglycosidases could bring the opportunity of designing novel therapeutics by targeting these important enzymes.
Collapse
Affiliation(s)
- Zhengliang L Wu
- Bio-techne, R&D Systems, Inc., 614 McKinley Place N.E., Minneapolis, MN 55413, USA
| | - James M Ertelt
- Bio-techne, R&D Systems, Inc., 614 McKinley Place N.E., Minneapolis, MN 55413, USA
| |
Collapse
|
36
|
Kotla NG, Bonam SR, Rasala S, Wankar J, Bohara RA, Bayry J, Rochev Y, Pandit A. Recent advances and prospects of hyaluronan as a multifunctional therapeutic system. J Control Release 2021; 336:598-620. [PMID: 34237401 DOI: 10.1016/j.jconrel.2021.07.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 12/12/2022]
Abstract
Hyaluronan (HA) is a naturally occurring non-sulfated glycosaminoglycan (GAG), cell-surface-associated biopolymer and is the key component of tissue extracellular matrix (ECM). Along with remarkable physicochemical properties, HA also has multifaceted biological effects that include but not limited to ECM organization, immunomodulation, and various cellular processes. Environmental cues such as tissue injury, infection or cancer change downstream signaling functionalities of HA. Unlike native HA, the fragments of HA have diversified effects on inflammation, cancer, fibrosis, angiogenesis and autoimmune response. In this review, we aim to discuss HA as a therapeutic delivery system development process, source, biophysical-chemical properties, and associated biological pathways (especially via cell surface receptors) of native and fragmented HA. We also tried to address an overview of the potential role of HA (native HA vs fragments) in the modulation of inflammation, immune response and various cancer targeting delivery applications. This review will also highlight the HA based therapeutic systems, medical devices and future perspectives of various biomedical applications were discussed in detail.
Collapse
Affiliation(s)
- Niranjan G Kotla
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, H91 W2TY, Ireland
| | - Srinivasa Reddy Bonam
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Equipe- Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, Université de Paris, Paris F-75006, France
| | - Swetha Rasala
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, H91 W2TY, Ireland
| | - Jitendra Wankar
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, H91 W2TY, Ireland
| | - Raghvendra A Bohara
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, H91 W2TY, Ireland
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Equipe- Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, Université de Paris, Paris F-75006, France; Indian Institute of Technology Palakkad, Palakkad 678 623, Kerala, India
| | - Yury Rochev
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, H91 W2TY, Ireland; Sechenov First Moscow State Medical University, Institute for Regenerative Medicine, Moscow 119992, Russia.
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, H91 W2TY, Ireland.
| |
Collapse
|
37
|
Goswami I, Bielitz R, Verbridge SS, von Spakovsky MR. A thermodynamic scaling law for electrically perturbed lipid membranes: Validation with steepest entropy ascent framework. Bioelectrochemistry 2021; 140:107800. [PMID: 33910115 DOI: 10.1016/j.bioelechem.2021.107800] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/20/2021] [Accepted: 03/09/2021] [Indexed: 10/21/2022]
Abstract
Experimental evidence has demonstrated the ability of transient pulses of electric fields to alter mammalian cell behavior. Strategies with these pulsed electric fields (PEFs) have been developed for clinical applications in cancer therapeutics, in-vivo decellularization, and tissue regeneration. Successful implementation of these strategies involve understanding how PEFs impact the cellular structures and, hence, cell behavior. The caveat, however, is that the PEF parameter space (i.e., comprising different pulse widths, amplitudes, number of pulses) is large, and design of experiments to explore all possible combinations of pulse parameters is prohibitive from a cost and time standpoint. In this study, a scaling law based on the Ising model is introduced to understand the impact of PEFs on the outer cell lipid membrane so that an understanding developed in one PEF pulse regime may be extended to another. Combining non-Markovian Monte Carlo techniques to determine density-of-states with a novel non-equilibrium thermodynamic framework based on the principle of steepest entropy ascent, the applicability of this scaling model to predict the behavior of both thermally quenched and electrically perturbed lipid membranes is demonstrated. A comparison of the predictions made by the steepest-entropy-ascent quantum thermodynamic (SEAQT) framework to experimental data is performed to validate the robustness of this computational methodology and the resulting scaling law.
Collapse
Affiliation(s)
- Ishan Goswami
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA 94720, USA; Department of Material Science and Engineering, University of California, Berkeley, CA 94720, USA.
| | - Robert Bielitz
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA
| | - Scott S Verbridge
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA
| | | |
Collapse
|
38
|
MCT4 Promotes Tumor Malignancy in F98 Glioma Cells. JOURNAL OF ONCOLOGY 2021; 2021:6655529. [PMID: 33936203 PMCID: PMC8060090 DOI: 10.1155/2021/6655529] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 03/16/2021] [Accepted: 03/25/2021] [Indexed: 12/31/2022]
Abstract
Monocarboxylate transporter 4 (MCT4, SLC16A3) is elevated under hypoxic conditions in many malignant tumors including gliomas. Moreover, MCT4 expression is associated with shorter overall survival. However, the functional consequences of MCT4 expression on the distinct hallmarks of cancer have not yet been explored at the cellular level. Here, we investigated the impact of MCT4 overexpression on proliferation, survival, cell death, migration, invasion, and angiogenesis in F98 glioma cells. Stable F98 glioma cell lines with MCT4 overexpression, normal expression, and knockdown were generated. Distinct hallmarks of cancer were examined using in silico analysis, various in vitro cell culture assays, and ex vivo organotypic rat brain slice culture model. Consistent with its function as lactate and proton exporter, MCT4 expression levels correlated inversely with extracellular pH and proportionally with extracellular lactate concentrations. Our results further indicate that MCT4 promotes proliferation and survival by altered cell cycle regulation and cell death mechanisms. Moreover, MCT4 overexpression enhances cell migration and invasiveness via reorganization of the actin cytoskeleton. Finally, MCT4 inhibition mitigates the induction of angiogenesis, suggesting that MCT4 also plays a crucial role in tumor-related angiogenesis. In summary, our data highlight MCT4/SLC16A3 as a key gene for distinct hallmarks of tumor malignancy in glioma cells.
Collapse
|
39
|
Koltai T, Reshkin SJ, Carvalho TMA, Cardone RA. Targeting the Stromal Pro-Tumoral Hyaluronan-CD44 Pathway in Pancreatic Cancer. Int J Mol Sci 2021; 22:3953. [PMID: 33921242 PMCID: PMC8069142 DOI: 10.3390/ijms22083953] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/01/2021] [Accepted: 04/08/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest malignancies. Present-day treatments have not shown real improvements in reducing the high mortality rate and the short survival of the disease. The average survival is less than 5% after 5 years. New innovative treatments are necessary to curtail the situation. The very dense pancreatic cancer stroma is a barrier that impedes the access of chemotherapeutic drugs and at the same time establishes a pro-proliferative symbiosis with the tumor, thus targeting the stroma has been suggested by many authors. No ideal drug or drug combination for this targeting has been found as yet. With this goal in mind, here we have explored a different complementary treatment based on abundant previous publications on repurposed drugs. The cell surface protein CD44 is the main receptor for hyaluronan binding. Many malignant tumors show over-expression/over-activity of both. This is particularly significant in pancreatic cancer. The independent inhibition of hyaluronan-producing cells, hyaluronan synthesis, and/or CD44 expression, has been found to decrease the tumor cell's proliferation, motility, invasion, and metastatic abilities. Targeting the hyaluronan-CD44 pathway seems to have been bypassed by conventional mainstream oncological practice. There are existing drugs that decrease the activity/expression of hyaluronan and CD44: 4-methylumbelliferone and bromelain respectively. Some drugs inhibit hyaluronan-producing cells such as pirfenidone. The association of these three drugs has never been tested either in the laboratory or in the clinical setting. We present a hypothesis, sustained by hard experimental evidence, suggesting that the simultaneous use of these nontoxic drugs can achieve synergistic or added effects in reducing invasion and metastatic potential, in PDAC. A non-toxic, low-cost scheme for inhibiting this pathway may offer an additional weapon for treating pancreatic cancer.
Collapse
Affiliation(s)
| | - Stephan Joel Reshkin
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (T.M.A.C.); (R.A.C.)
| | - Tiago M. A. Carvalho
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (T.M.A.C.); (R.A.C.)
| | - Rosa A. Cardone
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (T.M.A.C.); (R.A.C.)
| |
Collapse
|
40
|
Hu Y, Lou J, Jin Z, Yang X, Shan W, Du Q, Liao Q, Xu J, Xie R. Advances in research on the regulatory mechanism of NHE1 in tumors. Oncol Lett 2021; 21:273. [PMID: 33717270 PMCID: PMC7885159 DOI: 10.3892/ol.2021.12534] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/17/2020] [Indexed: 12/11/2022] Open
Abstract
Tumors pose a major threat to human health and present with difficulties that modern medicine has yet to overcome. It has been demonstrated that the acid-base balance of the tumor microenvironment is closely associated with the dynamic balance in the human body and that it regulates several processes, such as cell proliferation and differentiation, intracellular enzyme activity, and cytoskeletal assembly and depolymerization. It has been well established that the regulation of intra- and extracellular pH depends on a series of functional ion transporters and hydrogen ion channels, such as the Na+/H+ exchanger (NHE) protein and thee Cl/HCO3- exchange protein, among which the NHE1 member of the NHE family has been attracting increasing attention in recent years, particularly in studies on the correlation between pH regulation and tumors. NHE1 is a housekeeping gene encoding a protein that is widely expressed on the surface of all plasma membranes. Due to its functional domain, which determines the pHi at its N-terminus and C-terminus, NHE1 is involved in the regulation of the cellular pH microenvironment. It has been reported in the literature that NHE1 can regulate cell volume, participate in the transmembrane transport of intracellular and extracellular ions, affect cell proliferation and apoptosis, and regulate cell behavior and cell cycle progression; however, research on the role of NHE1 in tumorigenesis and tumor development in various systems is at its early stages. The aim of the present study was to review the current research on the correlation between the NHE family proteins and various systemic tumors, in order to indicate a new direction for antitumor drug development with the pH microenvironment as the target.
Collapse
Affiliation(s)
- Yanxia Hu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Jun Lou
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Zhe Jin
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Xiaoxu Yang
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Weixi Shan
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Qian Du
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Qiushi Liao
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Jingyu Xu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Rui Xie
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
41
|
Zheng Q, Zhang M, Zhou F, Zhang L, Meng X. The Breast Cancer Stem Cells Traits and Drug Resistance. Front Pharmacol 2021; 11:599965. [PMID: 33584277 PMCID: PMC7876385 DOI: 10.3389/fphar.2020.599965] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/17/2020] [Indexed: 12/13/2022] Open
Abstract
Drug resistance is a major challenge in breast cancer (BC) treatment at present. Accumulating studies indicate that breast cancer stem cells (BCSCs) are responsible for the BC drugs resistance, causing relapse and metastasis in BC patients. Thus, BCSCs elimination could reverse drug resistance and improve drug efficacy to benefit BC patients. Consequently, mastering the knowledge on the proliferation, resistance mechanisms, and separation of BCSCs in BC therapy is extremely helpful for BCSCs-targeted therapeutic strategies. Herein, we summarize the principal BCSCs surface markers and signaling pathways, and list the BCSCs-related drug resistance mechanisms in chemotherapy (CT), endocrine therapy (ET), and targeted therapy (TT), and display therapeutic strategies for targeting BCSCs to reverse drug resistance in BC. Even more importantly, more attention should be paid to studies on BCSC-targeted strategies to overcome the drug resistant dilemma of clinical therapies in the future.
Collapse
Affiliation(s)
- Qinghui Zheng
- Department of Breast Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Mengdi Zhang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Fangfang Zhou
- Institutes of Biology and Medical Science, Soochow University, Suzhou, China
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Xuli Meng
- Department of Breast Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China
| |
Collapse
|
42
|
Hozhabri H, Lashkari A, Razavi SM, Mohammadian A. Integration of gene expression data identifies key genes and pathways in colorectal cancer. Med Oncol 2021; 38:7. [PMID: 33411100 DOI: 10.1007/s12032-020-01448-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 11/21/2020] [Indexed: 12/16/2022]
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumor and prevalent cause of cancer-related death worldwide. In this study, we analyzed the gene expression profiles of patients with CRC with the aim of better understanding the molecular mechanism and key genes in CRC. Four gene expression profiles including, GSE9348, GSE41328, GSE41657, and GSE113513 were downloaded from GEO database. The data were processed using R programming language, in which 319 common differentially expressed genes including 94 up-regulated and 225 down-regulated were identified. The gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes pathway (KEGG) enrichment analyses were conducted to find the most significant enriched pathways in CRC. Based on the GO and KEGG pathway analysis, the most important dysregulated pathways were regulation of cell proliferation, biocarbonate transport, Wnt, and IL-17 signaling pathways, and nitrogen metabolism. The protein-protein interaction (PPI) network of the DEGs was constructed using Cytoscape software and hub genes including MYC, CXCL1, CD44, MMP1, and CXCL12 were identified as the most critical hub genes. The present study enhances our understanding of the molecular mechanisms of the CRC, which might potentially be applied in the treatment strategies of CRC as molecular targets and diagnostic biomarkers.
Collapse
Affiliation(s)
- Hossein Hozhabri
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| | - Ali Lashkari
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Seyed-Morteza Razavi
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.,Salari Institute of Cognitive and Behavioral Disorders (SICBD), Karaj, Alborz, Iran.,Systems Biology Research Lab, Bioinformatics Group, Systems Biology of Next Generation Company (SBNGC), Qom, Iran
| | - Ali Mohammadian
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
43
|
Mazloomi SM, Foroutan-Ghaznavi M, Montazeri V, Tavoosidana G, Fakhrjou A, Nozad-Charoudeh H, Pirouzpanah S. Profiling the expression of pro-metastatic genes in association with the clinicopathological features of primary breast cancer. Cancer Cell Int 2021; 21:6. [PMID: 33407452 PMCID: PMC7789694 DOI: 10.1186/s12935-020-01708-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/07/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Metastasis accounts for ninety percent of breast cancer (BrCa) mortality. Cortactin, Ras homologous gene family member A (RhoA), and Rho-associated kinase (ROCK) raise cellular motility in favor of metastasis. Claudins (CLDN) belong to tight junction integrity and are dysregulated in BrCa. Thus far, epidemiologic evidence regarding the association of different pro-metastatic genes with pathological phenotypes of BrCa is largely inconsistent. This study aimed to determine the possible transcriptional models of pro-metastatic genes incorporate in holding the integrity of epithelial cell-cell junctions (CTTN, RhoA, ROCK, CLDN-1, CLDN-2, and CLDN-4), for the first time, in association with clinicopathological features of primary BrCa. METHODS In a consecutive case-series design, 206 newly diagnosed non-metastatic eligible BrCa patients with histopathological confirmation (30-65 years) were recruited in Tabriz, Iran (2015-2017). Real-time RT-PCR was used. Then fold changes in the expression of target genes were measured. RESULTS ROCK amplification was associated with the involvement of axillary lymph node metastasis (ALNM; ORadj. = 3.05, 95%CI 1.01-9.18). Consistently, inter-correlations of CTTN-ROCK (β = 0.226, P < 0.05) and RhoA-ROCK (β = 0.311, P < 0.01) were determined among patients diagnosed with ALNM+ BrCa. In addition, the overexpression of CLDN-4 was frequently observed in tumors identified by ALNM+ or grade III (P < 0.05). The overexpression of CTTN, CLDN-1, and CLDN-4 genes was correlated positively with the extent of tumor size. CTTN overexpression was associated with the increased chance of luminal-A positivity vs. non-luminal-A (ORadj. = 1.96, 95%CI 1.02-3.77). ROCK was also expressed in luminal-B BrCa tumors (P < 0.05). The estrogen receptor-dependent transcriptions were extended to the inter-correlations of RhoA-ROCK (β = 0.280, P < 0.01), ROCK-CLDN-2 (β = 0.267, P < 0.05), and CLDN-1-CLDN-4 (β = 0.451, P < 0.001). CONCLUSIONS For the first time, our findings suggested that the inter-correlations of CTTN-ROCK and RhoA-ROCK were significant transcriptional profiles determined in association with ALNM involvement; therefore the overexpression of ROCK may serve as a potential molecular marker for lymphatic metastasis. The provided binary transcriptional profiles need more approvals in different clinical features of BrCa metastasis.
Collapse
Affiliation(s)
- Seyed-Mohammad Mazloomi
- Nutrition Research Center, Department of Food Hygiene and Quality Control, Faculty of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, 7193635899 Iran
| | - Mitra Foroutan-Ghaznavi
- Students’ Research Committee, Faculty of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, 7134814336 Iran
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, 5166414766 Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, 5166614756 Iran
| | - Vahid Montazeri
- Department of Thoracic Surgery, Faculty of Medicine, Tabriz University of Medical Sciences, Surgery Ward, Nour-Nejat Hospital, Tabriz, 5166614766 Iran
| | - Gholamreza Tavoosidana
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469 Iran
| | - Ashraf Fakhrjou
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, 5166614766 Iran
| | | | - Saeed Pirouzpanah
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, 5166414766 Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, 5166614756 Iran
- Department of Biochemistry and Dietetics, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, 5166614711 Iran
| |
Collapse
|
44
|
Hyaluronic acid and chondroitin sulfate (meth)acrylate-based hydrogels for tissue engineering: Synthesis, characteristics and pre-clinical evaluation. Biomaterials 2020; 268:120602. [PMID: 33360302 DOI: 10.1016/j.biomaterials.2020.120602] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/20/2022]
Abstract
Hydrogels based on photocrosslinkable Hyaluronic Acid Methacrylate (HAMA) and Chondroitin Sulfate Methacrylate (CSMA) are presently under investigation for tissue engineering applications. HAMA and CSMA gels offer tunable characteristics such as tailorable mechanical properties, swelling characteristics, and enzymatic degradability. This review gives an overview of the scientific literature published regarding the pre-clinical development of covalently crosslinked hydrogels that (partially) are based on HAMA and/or CSMA. Throughout the review, recommendations for the next steps in clinical translation of hydrogels based on HAMA or CSMA are made and potential pitfalls are defined. Specifically, a myriad of different synthetic routes to obtain polymerizable hyaluronic acid and chondroitin sulfate derivatives are described. The effects of important parameters such as degree of (meth)acrylation and molecular weight of the synthesized polymers on the formed hydrogels are discussed and useful analytical techniques for their characterization are summarized. Furthermore, the characteristics of the formed hydrogels including their enzymatic degradability are discussed. Finally, a summary of several recent applications of these hydrogels in applied fields such as cartilage and cardiac regeneration and advanced tissue modelling is presented.
Collapse
|
45
|
Abstract
Enzymes are a class of protein that catalyze a wide range of chemical reactions, including the cleavage of specific peptide bonds. They are expressed in all cell types, play vital roles in tissue development and homeostasis, and in many diseases, such as cancer. Enzymatic activity is tightly controlled through the use of inactive pro-enzymes, endogenous inhibitors and spatial localization. Since the presence of specific enzymes is often correlated with biological processes, and these proteins can directly modify biomolecules, they are an ideal biological input for cell-responsive biomaterials. These materials include both natural and synthetic polymers, cross-linked hydrogels and self-assembled peptide nanostructures. Within these systems enzymatic activity has been used to induce biodegradation, release therapeutic agents and for disease diagnosis. As technological advancements increase our ability to quantify the expression and nanoscale organization of proteins in cells and tissues, as well as the synthesis of increasingly complex and well-defined biomaterials, enzyme-responsive biomaterials are poised to play vital roles in the future of biomedicine.
Collapse
Affiliation(s)
- E. Thomas Pashuck
- Department of Bioengineering, P.C. Rossin College of Engineering and Applied Science, Lehigh University Bethlehem Pennsylvania USA
| |
Collapse
|
46
|
Dominguez-Gutierrez PR, Kwenda EP, Donelan W, O'Malley P, Crispen PL, Kusmartsev S. Hyal2 Expression in Tumor-Associated Myeloid Cells Mediates Cancer-Related Inflammation in Bladder Cancer. Cancer Res 2020; 81:648-657. [PMID: 33239427 DOI: 10.1158/0008-5472.can-20-1144] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 11/17/2020] [Accepted: 11/20/2020] [Indexed: 11/16/2022]
Abstract
The increased presence of myeloid-derived suppressor cells (MDSC) and tumor-associated macrophages (TAM) in tumor tissue has been extensively reported. However, their role in the regulation of hyaluronan (HA) metabolism in the tumor microenvironment has not been established. Here we describe a novel function of tumor-associated myeloid cells related to the enhanced breakdown of extracellular HA in human bladder cancer tissue, leading to the accumulation of small HA fragments with molecular weight (MW) <20 kDa. Increased fragmentation of extracellular HA and accumulation of low molecular weight HA (LMW-HA) in tumor tissue was associated with elevated production of multiple inflammatory cytokines, chemokines, and angiogenic factors. The fragmentation of HA by myeloid cells was mediated by the membrane-bound enzyme hyaluronidase 2 (Hyal2). Increased numbers of Hyal2+CD11b+ myeloid cells were detected in the tumor tissue as well as in the peripheral blood of patients with bladder cancer. Coexpression of CD33 suggested that these cells belong to monocytic myeloid-derived suppressor cells. The HA-degrading function of Hyal2-expressing MDSCs could be enhanced by exposure to tumor-conditioned medium, and IL1β was identified as one of the factors involved in the stimulation of Hyal2 activity. CD44-mediated signaling played an important role in the regulation of HA-degrading activity of Hyal2-expressing myeloid cells, as the engagement of CD44 receptor with specific mAb triggered translocation of Hyal2 enzyme to the cellular surface and stimulated secretion of IL1β. Taken together, this work identifies Hyal2-expressing tumor-associated myeloid cells as key players in the accumulation of LMW-HA in the tumor microenvironment and cancer-related inflammation and angiogenesis. SIGNIFICANCE: This study identifies Hyal2-expressing tumor-associated myeloid cells of monocyte-macrophage lineage as contributors to hyaluronan degradation in bladder cancer tissue, leading to accumulation of inflammatory and proangiogenic low molecular weight hyaluronan fragments.
Collapse
Affiliation(s)
| | | | - William Donelan
- Department of Urology, University of Florida, Gainesville, Florida
| | - Padraic O'Malley
- Department of Urology, University of Florida, Gainesville, Florida
| | - Paul L Crispen
- Department of Urology, University of Florida, Gainesville, Florida
| | | |
Collapse
|
47
|
Lactate in the Tumor Microenvironment: An Essential Molecule in Cancer Progression and Treatment. Cancers (Basel) 2020; 12:cancers12113244. [PMID: 33153193 PMCID: PMC7693872 DOI: 10.3390/cancers12113244] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/16/2020] [Accepted: 10/28/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary The role of lactate in cancer described by Otto Warburg in 1927 states that cancer cells uptake high amount of glucose with a marked increase in lactate production, this is known as the “Warburg effect”. Since then lactate turn out to be a major signaling molecule in cancer progression. Its release from tumor cells is accompanied by acidification ranging from 6.3 to 6.9 in the tumor microenvironment (TME) which favors processes such as tumor promotion, angiogenesis, metastasis, tumor resistance and more importantly, immunosuppression which has been associated with a poor outcome. The goal of this review is to examine and discuss in deep detail the recent studies that address the role of lactate in all these cancerous processes. Lastly, we explore the efforts to target the lactate production and its transport as a promising approach for cancer therapeutics. Abstract Cancer is a complex disease that includes the reprogramming of metabolic pathways by malignant proliferating cells, including those affecting the tumor microenvironment (TME). The “TME concept” was introduced in recognition of the roles played by factors other than tumor cells in cancer progression. In response to the hypoxic or semi-hypoxic characteristic of the TME, cancer cells generate a large amount of lactate via the metabolism of glucose and glutamine. Export of this newly generated lactate by the tumor cells together with H+ prevents intracellular acidification but acidifies the TME. In recent years, the importance of lactate and acidosis in carcinogenesis has gained increasing attention, including the role of lactate as a tumor-promoting metabolite. Here we review the existing literature on lactate metabolism in tumor cells and the ability of extracellular lactate to direct the metabolic reprogramming of those cells. Studies demonstrating the roles of lactate in biological processes that drive or sustain carcinogenesis (tumor promotion, angiogenesis, metastasis and tumor resistance) and lactate’s role as an immunosuppressor that contributes to tumor evasion are also considered. Finally, we consider recent therapeutic efforts using available drugs directed at and interfering with lactate production and transport in cancer treatment.
Collapse
|
48
|
Diverse Roles for Hyaluronan and Hyaluronan Receptors in the Developing and Adult Nervous System. Int J Mol Sci 2020; 21:ijms21175988. [PMID: 32825309 PMCID: PMC7504301 DOI: 10.3390/ijms21175988] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 02/07/2023] Open
Abstract
Hyaluronic acid (HA) plays a vital role in the extracellular matrix of neural tissues. Originally thought to hydrate tissues and provide mechanical support, it is now clear that HA is also a complex signaling molecule that can regulate cell processes in the developing and adult nervous systems. Signaling properties are determined by molecular weight, bound proteins, and signal transduction through specific receptors. HA signaling regulates processes such as proliferation, differentiation, migration, and process extension in a variety of cell types including neural stem cells, neurons, astrocytes, microglia, and oligodendrocyte progenitors. The synthesis and catabolism of HA and the expression of HA receptors are altered in disease and influence neuroinflammation and disease pathogenesis. This review discusses the roles of HA, its synthesis and breakdown, as well as receptor expression in neurodevelopment, nervous system function and disease.
Collapse
|
49
|
Riaz S, Abdulrahman N, Uddin S, Jabeen A, Gadeau AP, Fliegel L, Mraiche F. Anti-hypertrophic effect of Na +/H + exchanger-1 inhibition is mediated by reduced cathepsin B. Eur J Pharmacol 2020; 888:173420. [PMID: 32781168 DOI: 10.1016/j.ejphar.2020.173420] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/24/2020] [Accepted: 07/24/2020] [Indexed: 12/20/2022]
Abstract
Previous studies have established the role of Na+/H+ exchanger isoform-1 (NHE1) and cathepsin B (Cat B) in the development of cardiomyocyte hypertrophy (CH). Both NHE1 and Cat B are activated under acidic conditions suggesting that their activities might be interrelated. The inhibition of NHE1 has been demonstrated to reduce cardiac hypertrophy but the mechanism that contributes to the anti-hypertrophic effect of NHE1 inhibition still remains unclear. H9c2 cardiomyoblasts were stimulated with Angiotensin (Ang) II in the presence and absence of N-[2-methyl-4,5-bis(methylsulphonyl)-benzoyl]-guanidine, hydrochloride (EMD, EMD 87580), an NHE1 inhibitor or CA-074Me, a Cat B inhibitor, and various cardiac hypertrophic parameters, namely cell surface area, protein content and atrial natriuretic peptide (ANP) mRNA were analyzed. EMD significantly suppressed markers of cardiomyocyte hypertrophy and inhibited Ang II stimulated Cat B protein and gene expression. Cat B is located within the acidic environment of lysosomes. Cat B proteases are released into the cytoplasm upon disintegration of the lysosomes. EMD or CA-074Me prevented the dispersal of the lysosomes induced by Ang II and reduced the ratio of LC3-II to LC3-I, a marker of autophagy. Moreover, Cat B protein expression and MMP-9 activity in the extracellular space were significantly attenuated in the presence of EMD or CA-074Me. Our study demonstrates a novel mechanism for attenuation of the hypertrophic phenotype by NHE1 inhibition that is mediated by a regression in Cat B. The inhibition of Cat B via EMD or CA-074Me attenuates the autosomal-lysosomal pathway and MMP-9 activation.
Collapse
Affiliation(s)
- Sadaf Riaz
- College of Pharmacy, QU Health, Qatar University, Doha, Qatar; Hamad Medical Corporation, Doha, Qatar
| | - Nabeel Abdulrahman
- College of Pharmacy, QU Health, Qatar University, Doha, Qatar; Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Ayesha Jabeen
- College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | | | | | - Fatima Mraiche
- College of Pharmacy, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
50
|
Tang Y, Chen M, Xie Q, Li L, Zhu L, Ma Q, Gao S. Construction and evaluation of hyaluronic acid-based copolymers as a targeted chemotherapy drug carrier for cancer therapy. NANOTECHNOLOGY 2020; 31:305702. [PMID: 32272454 DOI: 10.1088/1361-6528/ab884d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Melanoma (MM) is a highly aggressive skin cancer with limited treatment options. Although chemotherapy has been using for advanced melanoma treatment, the lack of targetability, the poor biocompatibility and the severe side effects still hamper the wide applications of chemotherapy agents in MM management. Herein, a biocompatible and biodegradable polymeric hyaluronic acid nanoparticle (HANP) encapsulated with Paclitaxel (PTX) was developed for MM targeted therapy. Our results showed that PTX at 37 ± 2.1% (w/w) was able to be loaded into HANP with over 5 d of stability under physiological conditions. In vitro, HANP/PTX presented hyaluronidase-dependent drug release. Compared to free PTX, HANP/PTX demonstrated a 6-75 times higher growth inhibition in five different cancer cells, while only presenting minimum toxicity to normal cells. After intravenous administration at a 10 mg kg-1 equivalent dose of PTX, HANP/PTX significantly ablated MM tumor growth in a mouse model. As confirmed by 18F-fluoro-2-deoxy-D-glucose (FDG) positron emission tomography (PET) imaging, the tumors started to respond to the HANP/PTX as early as 7 d after the initial treatment, which will significantly benefit for personalized treatment. In conclusion, the HANP/PTX nanocomplex demonstrated great promise as a translational nanomedicine for cancer chemotherapy.
Collapse
Affiliation(s)
- Yuting Tang
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|