51
|
Wang J, Jordan AR, Zhu H, Hasanali SL, Thomas E, Lokeshwar SD, Morera DS, Alexander S, McDaniels J, Sharma A, Aguilar K, Sarcan S, Zhu T, Soloway MS, Terris MK, Thangaraju M, Lopez LE, Lokeshwar VB. Targeting hyaluronic acid synthase-3 (HAS3) for the treatment of advanced renal cell carcinoma. Cancer Cell Int 2022; 22:421. [PMID: 36581895 PMCID: PMC9801563 DOI: 10.1186/s12935-022-02818-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 11/30/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Hyaluronic acid (HA) promotes cancer metastasis; however, the currently approved treatments do not target HA. Metastatic renal carcinoma (mRCC) is an incurable disease. Sorafenib (SF) is a modestly effective antiangiogenic drug for mRCC. Although only endothelial cells express known SF targets, SF is cytotoxic to RCC cells at concentrations higher than the pharmacological-dose (5-µM). Using patient cohorts, mRCC models, and SF combination with 4-methylumbelliferone (MU), we discovered an SF target in RCC cells and targeted it for treatment. METHODS We analyzed HA-synthase (HAS1, HAS2, HAS3) expression in RCC cells and clinical (n = 129), TCGA-KIRC (n = 542), and TCGA-KIRP (n = 291) cohorts. We evaluated the efficacy of SF and SF plus MU combination in RCC cells, HAS3-transfectants, endothelial-RCC co-cultures, and xenografts. RESULTS RCC cells showed increased HAS3 expression. In the clinical and TCGA-KIRC/TCGA-KIRP cohorts, higher HAS3 levels predicted metastasis and shorter survival. At > 10-µM dose, SF inhibited HAS3/HA-synthesis and RCC cell growth. However, at ≤ 5-µM dose SF in combination with MU inhibited HAS3/HA synthesis, growth of RCC cells and endothelial-RCC co-cultures, and induced apoptosis. The combination inhibited motility/invasion and an HA-signaling-related invasive-signature. We previously showed that MU inhibits SF inactivation in RCC cells. While HAS3-knockdown transfectants were sensitive to SF, ectopic-HAS3-expression induced resistance to the combination. In RCC models, the combination inhibited tumor growth and metastasis with little toxicity; however, ectopic-HAS3-expressing tumors were resistant. CONCLUSION HAS3 is the first known target of SF in RCC cells. In combination with MU (human equivalent-dose, 0.6-1.1-g/day), SF targets HAS3 and effectively abrogates mRCC.
Collapse
Affiliation(s)
- Jiaojiao Wang
- grid.410427.40000 0001 2284 9329Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd, Room CN 1177A, Augusta, GA 30912 USA ,grid.513391.c0000 0004 8339 0314Present Address: Maoming People’s Hospital, Maoming, China
| | - Andre R. Jordan
- grid.410427.40000 0001 2284 9329Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd, Room CN 1177A, Augusta, GA 30912 USA ,grid.265219.b0000 0001 2217 8588Present Address: Tulane University School of Medicine, New Orleans, USA
| | - Huabin Zhu
- grid.410427.40000 0001 2284 9329Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd, Room CN 1177A, Augusta, GA 30912 USA ,grid.432444.1Present Address: Advanced RNA Technologies, Boulder, USA
| | - Sarrah L. Hasanali
- grid.410427.40000 0001 2284 9329Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd, Room CN 1177A, Augusta, GA 30912 USA ,grid.63368.380000 0004 0445 0041Present Address: Houston Methodist Hospital, Houston, USA
| | - Eric Thomas
- grid.410427.40000 0001 2284 9329Division of Urology, Department of Surgery, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd, Augusta, GA 30912 USA
| | - Soum D. Lokeshwar
- grid.410427.40000 0001 2284 9329Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd, Room CN 1177A, Augusta, GA 30912 USA ,grid.47100.320000000419368710Present Address: Yale University School of Medicine, New Haven, USA
| | - Daley S. Morera
- grid.410427.40000 0001 2284 9329Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd, Room CN 1177A, Augusta, GA 30912 USA
| | - Sung Alexander
- grid.410427.40000 0001 2284 9329Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd, Room CN 1177A, Augusta, GA 30912 USA
| | - Joseph McDaniels
- grid.410427.40000 0001 2284 9329Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd, Room CN 1177A, Augusta, GA 30912 USA
| | - Anuj Sharma
- grid.410427.40000 0001 2284 9329Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd, Room CN 1177A, Augusta, GA 30912 USA
| | - Karina Aguilar
- grid.410427.40000 0001 2284 9329Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd, Room CN 1177A, Augusta, GA 30912 USA
| | - Semih Sarcan
- grid.410427.40000 0001 2284 9329Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd, Room CN 1177A, Augusta, GA 30912 USA
| | - Tianyi Zhu
- Greenbrier High School, Evans, GA 30809 USA
| | - Mark S. Soloway
- grid.489080.d0000 0004 0444 4637Memorial Healthcare System, Aventura, FL 33180 USA
| | - Martha K. Terris
- grid.410427.40000 0001 2284 9329Division of Urology, Department of Surgery, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd, Augusta, GA 30912 USA
| | - Muthusamy Thangaraju
- grid.410427.40000 0001 2284 9329Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd, Room CN 1177A, Augusta, GA 30912 USA
| | - Luis E. Lopez
- grid.410427.40000 0001 2284 9329Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd, Room CN 1177A, Augusta, GA 30912 USA
| | - Vinata B. Lokeshwar
- grid.410427.40000 0001 2284 9329Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd, Room CN 1177A, Augusta, GA 30912 USA
| |
Collapse
|
52
|
Laomeephol C, Areecheewakul S, Tawinwung S, Suppipat K, Chunhacha P, Neves NM, Luckanagul JA. Potential roles of hyaluronic acid in in vivo CAR T cell reprogramming for cancer immunotherapy. NANOSCALE 2022; 14:17821-17840. [PMID: 36472072 DOI: 10.1039/d2nr05949e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has recently shown unprecedented clinical efficacy for cancer treatment, particularly of hematological malignancies. However, the complex manufacturing processes that involve ex vivo genetic modification of autologous T cells limits its therapeutic application. CAR T cells generated in vivo provide a valid alternative immunotherapy, "off-the-shelf", for cancer treatment. This approach requires carriers for the delivery of CAR-encoding constructs, which are plasmid DNA or messenger RNA, to T cells for CAR expression to help eradicate the tumor. As such, there are a growing number of studies reporting gene delivery systems for in vivo CAR T cell therapy based on viral vectors and polymeric nanoparticles. Hyaluronic acid (HA) is a natural biopolymer that can serve for gene delivery, because of its inherent properties of cell recognition and internalization, as well as its biodegradability, biocompatibility, and presence of functional groups for the chemical conjugation of targeting ligands. In this review, the potential of HA in the delivery of CAR constructs is discussed on the basis of previous experience of HA-based nanoparticles for gene therapy. Furthermore, current studies on CAR carriers for in vivo-generated CAR T cells are included, giving an idea of a rational design of HA-based systems for the more efficient delivery of CAR to circulating T cells.
Collapse
Affiliation(s)
- Chavee Laomeephol
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Sudartip Areecheewakul
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Supannikar Tawinwung
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Chulalongkorn University Cancer Immunology Excellence Center, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Koramit Suppipat
- Chulalongkorn University Cancer Immunology Excellence Center, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Chulalongkorn University Stem Cell and Cell Therapy Research Center, Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Preedakorn Chunhacha
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Nuno M Neves
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark - Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Jittima Amie Luckanagul
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
53
|
Hamdy NM, Eskander G, Basalious EB. Insights on the Dynamic Innovative Tumor Targeted-Nanoparticles-Based Drug Delivery Systems Activation Techniques. Int J Nanomedicine 2022; 17:6131-6155. [PMID: 36514378 PMCID: PMC9741821 DOI: 10.2147/ijn.s386037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
Anti-cancer conventional chemotherapeutic drugs novel formula progress, nowadays, uses nano technology for targeted drug delivery, specifically tailored to overcome therapeutic agents' delivery challenges. Polymer drug delivery systems (DDS) play a crucial role in minimizing off-target side effects arising when using standard cytotoxic drugs. Using nano-formula for targeted localized action, permits using larger effective cytotoxic doses on a single special spot, that can seriously cause harm if it was administered systemically. Therefore, various nanoparticles (NPs) specifically have attached groups for targeting capabilities, not seen in bulk materials, which then need activation. In this review, we will present a simple innovative, illustrative, in a cartoon-way, enumeration of NP anti-cancer drug targeting delivery system activation-types. Area(s) covered in this review are the mechanisms of various NP activation techniques.
Collapse
Affiliation(s)
- Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Georgette Eskander
- Faculty of Pharmacy, Ain Shams University, Postgraduate Student, Cairo, Egypt
| | - Emad B Basalious
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
54
|
Heydari M, Hosseinzadeh Colagar A, Moudi E. Mutant Allele of CD44 (rs8193C>T) and Pum2 Regulatory Element as A Prognosis Factor of Prostate Neoplasms: A Case-Control and In Silico Studies. CELL JOURNAL 2022; 24:723-731. [PMID: 36527344 PMCID: PMC9790067 DOI: 10.22074/cellj.2022.8468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Expression of CD44 variant 6 (CD44v6) as a homing-associated cell adhesion molecule (HCAM), has proved to change most cancer cells. Aim of the study is the effect of mutant allele of CD44 (rs8193C>T) and Pum2 regulatory element as a prognosis factor of prostate neoplasms: a case-control and in silico studies in the Mazandaran province-Iran. MATERIALS AND METHODS In a case-control study, CD44-rs8193C>T genotyping of the 420 prostate neoplasms (210 benign prostatic hyperplasia (BPH) patients and 210 prostate cancer patients) and 150 healthy samples are performed by the touchdown polymerase chain reaction with confronting two-pair primers (PCR-CTPP) method. The T mutant allele effects on the mRNA structure and cell pathways were also investigated in silico methods. RESULTS Our results showed that the increase of T mutant allele frequency was significantly associated with BPH compared with prostate cancer. Furthermore, results showed TT genotype was significantly associated with BPH [odds ratio (OR)=0.572 and P=0.015], and also influenced the CD44v6 transcript secondary structure, miRNA binding, and regulatory element-binding site for Pum2 protein. Attachment of Pum2 to standard CD44 transcript may lead to transcript isoform-switching and shift-expression to a variety of CD44 isoforms, which can trigger some of the cell signaling pathways, such as Nanog-Stat, PKC-Nanog, and PKC-Twist. CONCLUSION Based on this, the presence of the T mutant allele of CD44 (rs8193C>T) in the populations may create a regulatory element-binding site for Pum2. So, it could be known as a prognosis factor and prediction of prostate neoplasms. However, more comprehensive studies in different populations (with various ethnicities and large population sizes), and also CD44v6 gene expression studies in protein and transcript levels are required to confirm our data.
Collapse
Affiliation(s)
- Mohammadkazem Heydari
- Department of Molecular and Cell Biology, Faculty of Science, University of Mazandaran, Babolsar, Iran
| | - Abasalt Hosseinzadeh Colagar
- Department of Molecular and Cell Biology, Faculty of Science, University of Mazandaran, Babolsar, Iran,P.O.Box: 47416-95447Department of Molecular and Cell BiologyFaculty of ScienceUniversity of MazandaranBabolsarIran
| | - Emadoddin Moudi
- Department of Urology, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
55
|
Karalis T, Shiau AK, Gahman TC, Skandalis SS, Heldin CH, Heldin P. Identification of a Small Molecule Inhibitor of Hyaluronan Synthesis, DDIT, Targeting Breast Cancer Cells. Cancers (Basel) 2022; 14:cancers14235800. [PMID: 36497283 PMCID: PMC9741431 DOI: 10.3390/cancers14235800] [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: 10/10/2022] [Revised: 11/20/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022] Open
Abstract
Breast cancer is a common cancer in women. Breast cancer cells synthesize large amounts of hyaluronan to assist their proliferation, survival, migration and invasion. Accumulation of hyaluronan and overexpression of its receptor CD44 and hyaluronidase TMEM2 in breast tumors correlate with tumor progression and reduced overall survival of patients. Currently, the only known small molecule inhibitor of hyaluronan synthesis is 4-methyl-umbelliferone (4-MU). Due to the importance of hyaluronan for breast cancer progression, our aim was to identify new, potent and chemically distinct inhibitors of its synthesis. Here, we report a new small molecule inhibitor of hyaluronan synthesis, the thymidine analog 5'-Deoxy-5'-(1,3-Diphenyl-2-Imidazolidinyl)-Thymidine (DDIT). This compound is more potent than 4-MU and displays significant anti-tumorigenic properties. Specifically, DDIT inhibits breast cancer cell proliferation, migration, invasion and cancer stem cell self-renewal by suppressing HAS-synthesized hyaluronan. DDIT appears as a promising lead compound for the development of inhibitors of hyaluronan synthesis with potential usefulness in breast cancer treatment.
Collapse
Affiliation(s)
- Theodoros Karalis
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 572, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Andrew K. Shiau
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Timothy C. Gahman
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA
| | - Spyros S. Skandalis
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece
| | - Carl-Henrik Heldin
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 572, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Paraskevi Heldin
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 572, Uppsala University, SE-751 23 Uppsala, Sweden
- Correspondence: ; Tel.: +46-18-4714733
| |
Collapse
|
56
|
Pratt J, Haidara K, Annabi B. MT1-MMP Expression Levels and Catalytic Functions Dictate LDL Receptor-Related Protein-1 Ligand Internalization Capacity in U87 Glioblastoma Cells. Int J Mol Sci 2022; 23:ijms232214214. [PMID: 36430705 PMCID: PMC9692856 DOI: 10.3390/ijms232214214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
Modulations in cell surface receptor ectodomain proteolytic shedding impact on receptor function and cancer biomarker expression. As such, heavily pursued therapeutic avenues have exploited LDL receptor-related protein-1 (LRP-1)-mediated capacity in internalizing Angiopep-2 (An2), a brain-penetrating peptide that allows An2-drug conjugates to cross the blood-brain tumor barrier (BBTB). Given that LRP-1 is proteolytically shed from the cell surface through matrix metalloproteinase (MMP) activity, the balance between MMP expression/function and LRP-1-mediated An2 internalization is unknown. In this study, we found that membrane type-1 (MT1)-MMP expression increased from grade 1 to 4 brain tumors, while that of LRP-1 decreased inversely. MMP pharmacological inhibitors such as Ilomastat, Doxycycline and Actinonin increased in vitro An2 internalization by up to 2.5 fold within a human grade IV-derived U87 glioblastoma cell model. Transient siRNA-mediated MT1-MMP gene silencing resulted in increased basal An2 cell surface binding and intracellular uptake, while recombinant MT1-MMP overexpression reduced both cell surface LRP-1 expression as well as An2 internalization. The addition of Ilomastat to cells overexpressing recombinant MT1-MMP restored LRP-1 expression at the cell surface and An2 uptake to levels comparable to those observed in control cells. Collectively, our data suggest that MT1-MMP expression status dictates An2-mediated internalization processes in part by regulating cell surface LRP-1 functions. Such evidence prompts preclinical evaluations of combined MMP inhibitors/An2-drug conjugate administration to potentially increase the treatment of high-MT1-MMP-expressing brain tumors.
Collapse
|
57
|
Koletti AE, Kontogiannopoulos KN, Gardikis K, Letsiou S, Papageorgiou VP, Assimopoulou AN. Nanostructured lipid carriers of alkannins and shikonins: Experimental design, characterization and bioactivity studies. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
58
|
Xin Y, Sun Z, Liu J, Li W, Wang M, Chu Y, Sun Z, Deng G. Nanomaterial-mediated low-temperature photothermal therapy via heat shock protein inhibition. Front Bioeng Biotechnol 2022; 10:1027468. [PMID: 36304896 PMCID: PMC9595601 DOI: 10.3389/fbioe.2022.1027468] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 09/16/2022] [Indexed: 11/13/2022] Open
Abstract
With the continuous development of nanobiotechnology in recent years, combining photothermal materials with nanotechnology for tumor photothermal therapy (PTT) has drawn many attentions nanomedicine research. Although nanomaterial-mediated PTT is more specific and targeted than traditional treatment modalities, hyperthermia can also damage normal cells. Therefore, researchers have proposed the concept of low-temperature PTT, in which the expression of heat shock proteins (HSPs) is inhibited. In this article, the research strategies proposed in recent years based on the inhibition of HSPs expression to achieve low-temperature PTT was reviewed. Folowing this, the synthesis, properties, and applications of these nanomaterials were introduced. In addition, we also summarized the problems of nanomaterial-mediated low-temperature PTT at this stage and provided an outlook on future research directions.
Collapse
Affiliation(s)
- Yu Xin
- Yantai Yuhuangding Hospital, Yantai, China
| | - Zhuokai Sun
- Nanchang University Queen Mary School, Nanchang, China
| | - Jie Liu
- Yantai Yuhuangding Hospital, Yantai, China
| | - Wei Li
- Yantai Yuhuangding Hospital, Yantai, China
| | | | - Yongli Chu
- Yantai Yuhuangding Hospital, Yantai, China
| | - Zhihong Sun
- Yantai Yuhuangding Hospital, Yantai, China
- *Correspondence: Zhihong Sun, ; Guanjun Deng,
| | - Guanjun Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, China
- *Correspondence: Zhihong Sun, ; Guanjun Deng,
| |
Collapse
|
59
|
Babst N, Isbell LK, Rommel F, Tura A, Ranjbar M, Grisanti S, Tschuch C, Schueler J, Doostkam S, Reinacher PC, Duyster J, Kakkassery V, von Bubnoff N. CXCR4, CXCR5 and CD44 May Be Involved in Homing of Lymphoma Cells into the Eye in a Patient Derived Xenograft Homing Mouse Model for Primary Vitreoretinal Lymphoma. Int J Mol Sci 2022; 23:11757. [PMID: 36233057 PMCID: PMC9569795 DOI: 10.3390/ijms231911757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 09/25/2022] [Accepted: 09/30/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Primary vitreoretinal lymphoma (PVRL), a rare malignancy of the eye, is strongly related to primary central nervous system lymphoma (PCNSL). We hypothesized that lymphoma cells disseminate to the CNS and eye tissue via distinct homing receptors. The objective of this study was to test expression of CXCR4, CXCR5, CXCR7 and CD44 homing receptors on CD20 positive B-lymphoma cells on enucleated eyes using a PCNSL xenograft mouse model. Methods: We used indirect immunofluorescence double staining for CD20/CXCR4, CD20/CXCR5, CD20/CXCR7 and CD20/CD44 on enucleated eyes of a PCNSL xenograft mouse model with PVRL phenotype (PCNSL group) in comparison to a secondary CNS lymphoma xenograft mouse model (SCNSL group). Lymphoma infiltration was evaluated with an immunoreactive score (IRS). Results: 11/13 paired eyes of the PCNSL but none of the SCNSL group were infiltrated by CD20-positive cells. Particularly the choroid and to a lesser extent the retina of the PCNSL group were infiltrated by CD20+/CXCR4+, CD20+/CXCR5+, few CD20+/CD44+ but no CD20+/CXCR7+ cells. Expression of CXCR4 (p = 0.0205), CXCR5 (p = 0.0004) and CD44 (p < 0.0001) was significantly increased in the PCNSL compared to the SCNSL group. Conclusions: CD20+ PCNSL lymphoma cells infiltrating the eye co-express distinct homing receptors such as CXCR4 and CXCR5 in a PVRL homing mouse model. These receptors may be involved in PVRL homing into the eye.
Collapse
Affiliation(s)
- Neele Babst
- Department of Ophthalmology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Lisa K. Isbell
- Department of Medicine I, Medical Center—University of Freiburg, Faculty of Medicine, 79106 Freiburg, Germany
| | - Felix Rommel
- Department of Ophthalmology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Aysegul Tura
- Department of Ophthalmology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Mahdy Ranjbar
- Department of Ophthalmology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Salvatore Grisanti
- Department of Ophthalmology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Cordula Tschuch
- Charles River Discovery Research Services GmbH, 79108 Freiburg, Germany
| | - Julia Schueler
- Charles River Discovery Research Services GmbH, 79108 Freiburg, Germany
| | - Soroush Doostkam
- Institute for Neuropathology, Medical Center—University of Freiburg, Faculty of Medicine, 79106 Freiburg, Germany
| | - Peter C. Reinacher
- Department of Stereotactic and Functional Neurosurgery, Medical Center—University of Freiburg, Faculty of Medicine, 79106 Freiburg, Germany
- Fraunhofer Institute for Laser Technology (ILT), 52074 Aachen, Germany
| | - Justus Duyster
- Department of Medicine I, Medical Center—University of Freiburg, Faculty of Medicine, 79106 Freiburg, Germany
- German Cancer Consortium (DKTK), Freiburg, Germany and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Vinodh Kakkassery
- Department of Ophthalmology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Nikolas von Bubnoff
- Department of Hematology and Oncology, Medical Center, University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| |
Collapse
|
60
|
Mohammed S, Alhussien MN, Dang AK. Pregnancy stage-dependent modulation of neutrophil function may impact embryo survivability and pregnancy outcome in crossbred cows. Theriogenology 2022; 191:200-206. [PMID: 35998403 DOI: 10.1016/j.theriogenology.2022.08.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/26/2022]
Abstract
Pregnancy is a complicated physiological process that involves synchronized coordination between immune and endocrine systems. Neutrophils have been suggested as a critical immune cell for embryo implantation and pregnancy maintenance. The present study was conducted to evaluate the dynamic changes in the mRNA expressions of the cluster of designation (CD11b, CD31, CD44 and CD62L) molecules and interferon-stimulated genes (ISG15, MX1 and OAS1) in blood neutrophils throughout pregnancy in dairy cows and correlate them with the outcome of pregnancy. Blood samples were taken from negative control (NC) group, and non-pregnant (NP) group at the time of artificial insemination (AI, day zero) and on days 10, 14, 16, 18, and 21 post-AI. In pregnant (P) cows, samples were taken as described above and after every 30 days until the time of parturition. In aborted cows, samples were collected until the time of the abortion. Comparison between pregnant, non-pregnant and aborted cows revealed that the expression of CD molecules increased (p < 0.05) on days 14, 16, 18 and 21 post-AI only in NP cows as compared to other groups. Although the expression of CD molecules remained constant throughout the study period in pregnant and aborted cows, the expression of CD11b, CD31 and CD62L increased (p < 0.05) on the day of abortion and parturition. Unlike CD molecules, the expression of CD44 decreased significantly (p < 0.05) at the time of abortion. There was a significant (p < 0.05) increase in the expression of interferon-stimulated genes including MX1, OAS1 and ISG15 during the peri-implantation period in pregnant cows, and at the time of abortion in aborted cows. However, the expression of ISGs was lower (p < 0.05) in non-pregnant cows as compared to the other groups. The results revealed the critical role played by neutrophils during pregnancy and form the basis to unravel the underlying mechanism for neutrophil associated immunological infertility in bovines.
Collapse
Affiliation(s)
- Seid Mohammed
- Bio and Emerging Technology Institute, Addis Ababa, Ethiopia; Lactation and Immuno-Physiology Laboratory, ICAR-National Dairy Research Institute, Karnal, 132001, Haryana, India
| | - Mohanned Naif Alhussien
- Lactation and Immuno-Physiology Laboratory, ICAR-National Dairy Research Institute, Karnal, 132001, Haryana, India; Reproductive Biotechnology, TUM School of Life Sciences Weihenstephan, Technical University Munich, 85354, Freising, Germany.
| | - Ajay Kumar Dang
- Lactation and Immuno-Physiology Laboratory, ICAR-National Dairy Research Institute, Karnal, 132001, Haryana, India
| |
Collapse
|
61
|
Singh VK, Chau E, Mishra A, DeAnda A, Hegde VL, Sastry JK, Haviland D, Jagannath C, Godin B, Khan A. CD44 receptor targeted nanoparticles augment immunity against tuberculosis in mice. J Control Release 2022; 349:796-811. [PMID: 35914613 PMCID: PMC10478167 DOI: 10.1016/j.jconrel.2022.07.040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/26/2022] [Accepted: 07/26/2022] [Indexed: 02/01/2023]
Abstract
We describe a role of CD44-mediated signaling during host-defense against tuberculosis (TB) using a mouse model of TB and studies in M. tuberculosis (Mtb) infected human macrophage (MФ). Liposomes targeting CD44 using thioaptamers (CD44TA-LIP) were designed and tested as new vaccines to boost host immunity in TB. CD44TA-LIP enhanced killing of Mtb in human MФ, which correlated with an increased production of pro-inflammatory cytokines IL-1β, TNF-α and IL-12. CD44TA-LIP activated MФ showed an enhanced MHC-II dependent antigen presentation to CD4 T-cells. Inhibition of cellular proliferation and cytoskeleton rearrangement pathways downstream of CD44 signaling abrogated CD44TA-LIP-induced antimicrobial effects. Blockade of inflammatory pathways also reduced antigen presentation by MФ and activation of CD4 T cells. Mtb infected MФ treated with CD44TA-LIP exhibited increased nitric oxide and HβD2 defensin peptide production. Among Mtb infected mice with increased lung and spleen loads of organisms, intranasal administration of CD44TA-LIP led to a ten-fold reduction of colony forming units of Mtb and elevated IFN-γ + CD4, effector, central and resident memory T cells. Biodistribution studies demonstrated that CD44TA-LIP preferentially accumulated in the lungs and were associated with CD11b + cells. CD44TA-LIP treated mice showed no weight loss or increased liver LDH levels. This study highlights the importance of CD44-mediated signaling in host-defense during TB and the therapeutic potential of CD44TA-LIP.
Collapse
Affiliation(s)
- Vipul K Singh
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Eric Chau
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Abhishek Mishra
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Alexandro DeAnda
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Venkatesh L Hegde
- Department of Thoracic Head & Neck Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Jagannadha K Sastry
- Department of Thoracic Head & Neck Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - David Haviland
- Flow Cytometry Core, Houston Methodist Research Institute, Houston, TX, USA
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, TX, USA.
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA.
| | - Arshad Khan
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, TX, USA.
| |
Collapse
|
62
|
Cao L, Fang H, Yan D, Wu XM, Zhang J, Chang MX. CD44a functions as a regulator of p53 signaling, apoptosis and autophagy in the antibacterial immune response. Commun Biol 2022; 5:889. [PMID: 36042265 PMCID: PMC9427754 DOI: 10.1038/s42003-022-03856-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 08/17/2022] [Indexed: 11/26/2022] Open
Abstract
The cell adhesion molecule CD44 has been implicated in diverse biological functions including the pathological responses to infections and inflammatory diseases. The variable forms of CD44 contribute to functional variations, which are not yet defined in teleost. Here, we show that zebrafish CD44a plays a protective role in the host defense against Edwardsiella piscicida infection. Zebrafish CD44a deficiency inhibits cell growth and proliferation, impairs cell growth and death pathways, and regulates the expression levels of many genes involved in p53 signaling, apoptosis and autophagy. In addition, CD44a gene disruption in zebrafish leads to inhibition of apoptosis and induction of autophagy, with the increased susceptibility to E. piscicida infection. Furthermore, we show that zebrafish CD44a variants including CD44a_tv1 and CD44a_tv2 promote the translocation of p53 from the nucleus to the cytoplasm and interact with p53 in the cytoplasm. Mechanistically, zebrafish CD44a_tv1 mediates the beneficial effect for larvae survival infected with E. piscicida is depending on the CASP8-mediated apoptosis. However, the antibacterial effect of zebrafish CD44a_tv2 depends on the cytoplasmic p53-mediated inhibition of autophagy. Collectively, our results identify that different mechanisms regulate CD44a variants-mediated antibacterial responses.
Collapse
Affiliation(s)
- Lu Cao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Hong Fang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Dong Yan
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Xiao Man Wu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Jie Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Ming Xian Chang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China.
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, 100049, Beijing, China.
- Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China.
| |
Collapse
|
63
|
Soares DJ. Bridging a Century-Old Problem: The Pathophysiology and Molecular Mechanisms of HA Filler-Induced Vascular Occlusion (FIVO)-Implications for Therapeutic Interventions. Molecules 2022; 27:5398. [PMID: 36080164 PMCID: PMC9458226 DOI: 10.3390/molecules27175398] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 01/10/2023] Open
Abstract
Biocompatible hyaluronic acid (HA, hyaluronan) gel implants have altered the therapeutic landscape of surgery and medicine, fostering an array of innovative products that include viscosurgical aids, synovial supplements, and drug-eluting nanomaterials. However, it is perhaps the explosive growth in the cosmetic applications of injectable dermal fillers that has captured the brightest spotlight, emerging as the dominant modality in plastic surgery and aesthetic medicine. The popularity surge with which injectable HA fillers have risen to in vogue status has also brought a concomitant increase in the incidence of once-rare iatrogenic vaso-occlusive injuries ranging from disfiguring facial skin necrosis to disabling neuro-ophthalmological sequelae. As our understanding of the pathophysiology of these injuries has evolved, supplemented by more than a century of astute observations, the formulation of novel therapeutic and preventative strategies has permitted the amelioration of this burdensome complication. In this special issue article, we review the relevant mechanisms underlying HA filler-induced vascular occlusion (FIVO), with particular emphasis on the rheo-mechanical aspects of vascular blockade; the thromboembolic potential of HA mixtures; and the tissue-specific ischemic susceptibility of microvascular networks, which leads to underperfusion, hypoxia, and ultimate injury. In addition, recent therapeutic advances and novel considerations on the prevention and management of muco-cutaneous and neuro-ophthalmological complications are examined.
Collapse
Affiliation(s)
- Danny J. Soares
- American Foundation for Aesthetic Medicine (AFFAM), Fruitland Park, FL 34731, USA;
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| |
Collapse
|
64
|
Expression of CD44 in Leukocyte Subpopulations in Patients with Inflammatory Bowel Diseases. Diagnostics (Basel) 2022; 12:diagnostics12082014. [PMID: 36010364 PMCID: PMC9407096 DOI: 10.3390/diagnostics12082014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 11/17/2022] Open
Abstract
CD44 expressed in monocytes and lymphocytes seems to play a crucial role in gastrointestinal inflammation, such as the one occurring in the context of inflammatory bowel diseases. Differentially methylated genes are distinctly expressed across monocyte subpopulations related to the state of Crohn’s disease. Hence, the aim of this study was to detect CD44 expression in leukocyte subpopulations in relation to the type of IBD, therapy, and disease duration. Monocyte subpopulations CD14++CD16−, CD14++CD16++, and CD14+CD16+ as well as other leukocytes were analyzed for their CD44 expression using flow cytometry in 46 patients with IBD and 48 healthy controls. Patients with Crohn’s disease treated with non-biological therapy (NBT) exhibited a lower percentage of anti-inflammatory CD14+CD16++ monocytes, whereas NBT-treated patients with ulcerative colitis had lower expression of CD44 on CD14+CD44+ lymphocytes in comparison to controls, respectively. Conversely, patients with Crohn’s disease treated with biological therapy had a higher percentage of CD44+ granulocytes but lower expression of CD44 on anti-inflammatory monocytes compared to controls. Median fluorescence intensity (MFI) of CD44 on CD44+CD14+ lymphocytes was higher in ulcerative colitis patients treated with biological therapy compared to NBT. The percentage of classical CD14++CD16− monocytes was lower in the <9 years of IBD duration subgroup compared with the longer disease duration subgroup. The present study addresses the putative role of differentiation and regulation of leukocytes in tailoring IBD therapeutic regimes.
Collapse
|
65
|
Dartora VFC, Salata GC, Passos JS, Branco PC, Silveira E, Steiner AA, Costa-Lotufo LV, Lopes LB. Hyaluronic acid nanoemulsions improve piplartine cytotoxicity in 2D and 3D breast cancer models and reduce tumor development after intraductal administration. Int J Biol Macromol 2022; 219:84-95. [PMID: 35907458 DOI: 10.1016/j.ijbiomac.2022.07.162] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/13/2022] [Accepted: 07/20/2022] [Indexed: 12/24/2022]
Abstract
Nanoemulsions modified with chitosan (NE-Q) or hyaluronic acid (NE-HA), developed for intraductal administration of piplartine (piperlongumine) and local breast cancer treatment, were evaluated for cytotoxic effects in vitro in 2D and 3D breast cancer models and in vivo in a chemically induced carcinogenesis model. Droplet size was lower than 100 nm, and zeta potential varied from +17.9 to -25.5 mV for NE-Q and NE-HA, respectively. Piplartine nanoencapsulation reduced its IC50 up to 3.6-fold in T-47D and MCF-7 monolayers without differences between NE-Q and NE-HA, and up to 6.6-fold in cancer spheroids. Cytotoxicity improvement may result from a more efficient NE-mediated delivery, as suggested by stronger fluorescent staining of cells and spheroids. In 1-methyl-1-nitrosourea -induced breast cancer models, intraductal administration of piplartine-loaded NE-HA inhibited breast tumor development and histological alterations. These results support the potential applicability of piplartine-loaded NE-HA for intraductal treatment of breast cancer.
Collapse
Affiliation(s)
- Vanessa F C Dartora
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil; Departamento de Imunologia, Instituto de Ciências Biomédicas IV, São Paulo, SP, Brazil
| | - Giovanna C Salata
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Julia S Passos
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Paola C Branco
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | | | - Leticia V Costa-Lotufo
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Luciana B Lopes
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
66
|
Kang MS, Kwon M, Lee SH, Kim WH, Lee GW, Jo HJ, Kim B, Yang SY, Kim KS, Han DW. 3D printing of skin equivalents with hair follicle structures and epidermal-papillary-dermal layers using gelatin/hyaluronic acid hydrogels. Chem Asian J 2022; 17:e202200620. [PMID: 35866189 DOI: 10.1002/asia.202200620] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/18/2022] [Indexed: 11/10/2022]
Abstract
Recent advances in three-dimensional (3D) bioprinting technologies enabled the fabrication of sophisticated live 3D tissue analogs. Although various hydrogel-based bioink has been reported, the development of advanced bioink materials that can reproduce the composition of native extracellular matrix (ECM) accurately and mimic the intrinsic property of laden cells is still challenging. In this work, 3D printed skin equivalents incorporating hair follicle structures and epidermal-papillary-dermal layers are fabricated with gelatin methacryloyl (GelMA)/hyaluronic acid (HA) MA (HAMA) hydrogel (GelMA/HAMA) bioink. The composition of collagen and glycosaminoglycan (GAG) of native skin was recapitulated by adjusting the combination of GelMA and HAMA. The GelMA/HAMA bioink was proven to have excellent viscoelastic and physicochemical properties, 3D printability, cytocompatibility, and functionality to maintain the hair inductive potency and facilitated spontaneous hair pore development. Overall, we suggest that the GelMA/HAMA hydrogels can be promising candidates as bioinks for the 3D printing of skin equivalents with epidermal-papillary-dermal multi-layers and hair follicle structures, and they might serve as a useful model in skin tissue engineering and regeneration.
Collapse
Affiliation(s)
- Moon Sung Kang
- Pusan National University, Cogno-Mechatronics Engineering, KOREA, REPUBLIC OF
| | - Mina Kwon
- Pusan National University, School of Chemical Engineering, KOREA, REPUBLIC OF
| | - Seok Hyun Lee
- Pusan National University, Cogno-Mechatronics Engineering, KOREA, REPUBLIC OF
| | - Won-Hyeon Kim
- Seoul National University Dental Hospital, Dental Life Science Research Institute, KOREA, REPUBLIC OF
| | - Gyeong Won Lee
- Pusan National University - Milyang Campus, Biomaterials Science, KOREA, REPUBLIC OF
| | - Hyo Jung Jo
- Pusan National University, Cogno-Mechatronics Engineering, KOREA, REPUBLIC OF
| | - Bongju Kim
- Seoul National University Dental Hospital, Dental Life Science Research Institute, KOREA, REPUBLIC OF
| | - Seung Yun Yang
- Pusan National University - Milyang Campus, Biomaterials Science, KOREA, REPUBLIC OF
| | - Ki Su Kim
- Pusan National University, School of Chemical Engineering, KOREA, REPUBLIC OF
| | - Dong-Wook Han
- Pusan National University, Cogno-Mechatronics Engineering, Department of Cogno-Mechatronics Engineering, College of Nanoscience & Nanotechnology, Pusan National University, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Korea, 46241, Busan, KOREA, REPUBLIC OF
| |
Collapse
|
67
|
Krstić L, Jarho P, Ruponen M, Urtti A, González-García MJ, Diebold Y. Improved ocular delivery of quercetin and resveratrol: A comparative study between binary and ternary cyclodextrin complexes. Int J Pharm 2022; 624:122028. [PMID: 35853567 DOI: 10.1016/j.ijpharm.2022.122028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 11/19/2022]
Abstract
The number of patients affected by Dry Eye Disease (DED) had notably increased worldwide, addressing the need of novel therapeutic approaches. Polyphenols, quercetin (QUE) and resveratrol (RSV) show necessary antioxidant and anti-inflammatory properties to manage DED, but their application as topical eyedrops is restricted by low aqueous solubility and low chemical stability. Cyclodextrins (CD) are widely used to improve physicochemical characteristics of drugs. Consequently, the aim of this study was to make a comparison between binary complexes with quercetin, resveratrol and cyclodextrins and tertiary complexes adding hyaluronic acid (HA). Both complexes were able to enhance solubility and stability of QUE and RSV. AFM imaging and DLS measurements disclose the formation of spherical nanoaggregates within tertiary complexes of both QUE and RSV with mean diameters of 103 and 82 nm. Neither complex demonstrated cytotoxic effect in in vitro studies in corneal (HCE) and conjunctival (IM-ConjEpi) cell lines. In HCE cells, complexes containing QUE or RSV at their highest concentrations were able to scavenge more than 95 % of the ROS that were produced intracellularly (p < 0.005). Similar response was observed with IM-ConjEpi cells. The antioxidant effect was maintained in the complexes with HA. This confirmed their potential as viable topical treatment for DED.
Collapse
Affiliation(s)
- Luna Krstić
- Insituto de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, 47011 Valladolid, Spain
| | - Pekka Jarho
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1, 70210 Kuopio, Finland
| | - Marika Ruponen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1, 70210 Kuopio, Finland
| | - Arto Urtti
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1, 70210 Kuopio, Finland; Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00014 Helsinki, Finland
| | - María J González-García
- Insituto de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, 47011 Valladolid, Spain; Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Yolanda Diebold
- Insituto de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, 47011 Valladolid, Spain; Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
68
|
Ying W, Hu Y, Zhu H. Expression of CD44, Transforming Growth Factor-β, and Matrix Metalloproteinases in Women With Pelvic Organ Prolapse. Front Surg 2022; 9:902871. [PMID: 35910471 PMCID: PMC9334776 DOI: 10.3389/fsurg.2022.902871] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Background Defects in the pelvic floor connective tissue may underlie the etiology of pelvic organ prolapse (POP). We hypothesized that the expression of proteins regulating extracellular matrix turnover is altered in the uterosacral ligament of women with POP. We compared the expression of CD44, transforming growth factor (TGF)-β, and matrix metalloproteinases (MMPs) 2/9 in women with and without POP. Methods and Results This matched case-control study included 30 postmenopausal women, with POP stage 2 and higher according to the POP quantification system, and 30 postmenopausal women without POP. Immunohistochemical analyses of the uterosacral ligament specimens obtained after hysterectomy were performed to determine CD44, TGF-β, MMP-2, and MMP-9 expression. The expression was quantified using ImageJ software, and the association between prolapse occurrence and risk factors was evaluated using Spearman's correlation analysis. CD44 expressions were significantly lower (p < 0.05), whereas MMP-2 and MMP-9 expression was higher (p < 0.0001 and p < 0.05, respectively), in the POP group than in the control group. The expression of TGF-β was similar in both groups. The occurrence of uterine prolapse was positively correlated with age, postmenopausal age, and MMP-2 and MMP-9 expression (p < 0.01) and negatively correlated with CD44 expression (p < 0.05). Conclusion CD44, MMP-2, and MMP-9 may play critical roles in the pathogenesis of POP and may be candidate biomarkers of POP progression.
Collapse
Affiliation(s)
- Weiwei Ying
- Department of Gynecology, Taizhou Hospital of Zhejiang Province, Zhejiang University, Taizhou, China
| | - Yanping Hu
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, HangZhou, China
| | - Haibin Zhu
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, HangZhou, China
- Correspondence: HaiBin Zhu
| |
Collapse
|
69
|
Overview of Transcriptomic Research on Type 2 Diabetes: Challenges and Perspectives. Genes (Basel) 2022; 13:genes13071176. [PMID: 35885959 PMCID: PMC9319211 DOI: 10.3390/genes13071176] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 02/04/2023] Open
Abstract
Type 2 diabetes (T2D) is a common chronic disease whose etiology is known to have a strong genetic component. Standard genetic approaches, although allowing for the detection of a number of gene variants associated with the disease as well as differentially expressed genes, cannot fully explain the hereditary factor in T2D. The explosive growth in the genomic sequencing technologies over the last decades provided an exceptional impetus for transcriptomic studies and new approaches to gene expression measurement, such as RNA-sequencing (RNA-seq) and single-cell technologies. The transcriptomic analysis has the potential to find new biomarkers to identify risk groups for developing T2D and its microvascular and macrovascular complications, which will significantly affect the strategies for early diagnosis, treatment, and preventing the development of complications. In this article, we focused on transcriptomic studies conducted using expression arrays, RNA-seq, and single-cell sequencing to highlight recent findings related to T2D and challenges associated with transcriptome experiments.
Collapse
|
70
|
González-Sánchez E, Muñoz-Callejas A, Gómez-Román J, San Antonio E, Marengo A, Tsapis N, Bohne-Japiassu K, González-Tajuelo R, Pereda S, García-Pérez J, Cavagna L, González-Gay MÁ, Vicente-Rabaneda E, Meloni F, Fattal E, Castañeda S, Urzainqui A. Everolimus targeted nanotherapy reduces inflammation and fibrosis in scleroderma-related interstitial lung disease (SSc-ILD) developed by PSGL-1 deficient mice. Br J Pharmacol 2022; 179:4534-4548. [PMID: 35726496 DOI: 10.1111/bph.15898] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 02/21/2022] [Accepted: 04/11/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Interstitial lung disease (ILD) is the main cause of mortality in systemic sclerosis (SSc) and current therapies available are of low efficacy or high toxicity. Thus, the identification of innovative less toxic and high efficacy therapeutic approaches to ILD treatment is a crucial point. P-selectin Glycoprotein Ligand-1 (PSGL-1) interaction with P-selectin initiates leukocyte extravasation and the lack of its expression brings to SSc-like syndrome with high incidence of ILD in aged mice. EXPERIMENTAL APPROACH Aged PSGL-1-/- mice were used to assay the therapeutic efficacy of an innovative nanotherapy with everolimus (Ev), included in liposomes decorated with high MW hyaluronic acid (LipHA+Ev) and administrated intratracheally to specifically target CD44-expressing lung cells. KEY RESULTS PSGL-1-/- mice had increased number of CD45+ and CD45- cells, including alveolar and interstitial macrophages, eosinophils, granulocytes and NK cells, and elevated number of myofibroblasts in broncoalveolar lavage (BAL). CD45+ and CD45- cells expressing proinflammatory and profibrotic cytokines were also increased. PSGL-1-/- mice lung histopathology showed increased immune cell infiltration and apoptosis and exacerbated interstitial and peribronchial fibrosis. Targeted nanotherapy with LipHA+Ev reduced BAL number of myofibroblast, cells producing proinflammatory and profibrotic cytokines, and the degree of lung inflammation at histology. LipHA+Ev treatment also provided an important decrease in severity of peribronchial and interstitial lung fibrosis from moderate to mild injury score. CONCLUSIONS AND IMPLICATIONS Our preclinical study in PSGL-1-/- mice indicates that targeted nanotherapy with LipHA+Ev represents an effective treatment for SSc-ILD, reducing the number of inflammatory and fibrotic cells in BAL and reducing inflammation and fibrosis in lungs.
Collapse
Affiliation(s)
- Elena González-Sánchez
- Immunology Department, Hospital Universitario de la Princesa, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Madrid, Spain
| | - Antonio Muñoz-Callejas
- Immunology Department, Hospital Universitario de la Princesa, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Madrid, Spain
| | - Javier Gómez-Román
- Pathology Department, Hospital Universitario Marqués de Valdecilla, IDIVAL, Universidad de Cantabria, Santander, Cantabria, Spain
| | - Esther San Antonio
- Immunology Department, Hospital Universitario de la Princesa, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Madrid, Spain
| | - Alessandro Marengo
- Institut Galien Paris Sud, UMR CNRS 8612. School of Pharmacy at University Paris-Saclay, Châtenay-Malabry, France
| | - Nicolas Tsapis
- Institut Galien Paris Sud, UMR CNRS 8612. School of Pharmacy at University Paris-Saclay, Châtenay-Malabry, France
| | - Kamila Bohne-Japiassu
- Institut Galien Paris Sud, UMR CNRS 8612. School of Pharmacy at University Paris-Saclay, Châtenay-Malabry, France
| | - Rafael González-Tajuelo
- Immunology Department, Hospital Universitario de la Princesa, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Madrid, Spain
| | - Saray Pereda
- Pathology Department, Hospital Universitario Marqués de Valdecilla, IDIVAL, Universidad de Cantabria, Santander, Cantabria, Spain
| | - Javier García-Pérez
- Pneumology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital Universitario de la Princesa, Madrid, Spain
| | - Lorenzo Cavagna
- Rheumatology Department, University and IRCCS Policlinico S. Matteo Foundation, Università degli Studi di Pavia, Pavia, Italy
| | - Miguel Ángel González-Gay
- Rheumatology Department, Hospital Universitario Marqués de Valdecilla, IDIVAL, Universidad de Cantabria, Santander, Spain
| | - Esther Vicente-Rabaneda
- Rheumatology Department, Hospital Universitario de la Princesa, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Madrid, Spain
| | - Federica Meloni
- Internal Medicine Department, Pneumology Division, IRCCS San Matteo Foundation and Università degli Studi di Pavia, Pavia, Italy
| | - Elias Fattal
- Institut Galien Paris Sud, UMR CNRS 8612. School of Pharmacy at University Paris-Saclay, Châtenay-Malabry, France
| | - Santos Castañeda
- Rheumatology Department, Hospital Universitario de la Princesa, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Madrid, Spain.,Cathedra UAM-Roche, EPID-Future, Department of Medicine, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Ana Urzainqui
- Immunology Department, Hospital Universitario de la Princesa, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Madrid, Spain
| |
Collapse
|
71
|
Chondroitin Sulfate: Emerging biomaterial for biopharmaceutical purpose and tissue engineering. Carbohydr Polym 2022; 286:119305. [PMID: 35337491 DOI: 10.1016/j.carbpol.2022.119305] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/28/2022] [Accepted: 02/28/2022] [Indexed: 12/20/2022]
|
72
|
Gairola A, Benjamin A, Weatherston JD, Cirillo JD, Wu HJ. Recent Developments in Drug Delivery for Treatment of Tuberculosis by Targeting Macrophages. ADVANCED THERAPEUTICS 2022; 5:2100193. [PMID: 36203881 PMCID: PMC9531895 DOI: 10.1002/adtp.202100193] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Indexed: 11/10/2022]
Abstract
Tuberculosis (TB) is among the greatest public health and safety concerns in the 21st century, Mycobacterium tuberculosis, which causes TB, infects alveolar macrophages and uses these cells as one of its primary sites of replication. The current TB treatment regimen, which consist of chemotherapy involving a combination of 3-4 antimicrobials for a duration of 6-12 months, is marked with significant side effects, toxicity, and poor compliance. Targeted drug delivery offers a strategy that could overcome many of the problems of current TB treatment by specifically targeting infected macrophages. Recent advances in nanotechnology and material science have opened an avenue to explore drug carriers that actively and passively target macrophages. This approach can increase the drug penetration into macrophages by using ligands on the nanocarrier that interact with specific receptors for macrophages. This review encompasses the recent development of drug carriers specifically targeting macrophages actively and passively. Future directions and challenges associated with development of effective TB treatment is also discussed.
Collapse
Affiliation(s)
- Anirudh Gairola
- Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA
| | - Aaron Benjamin
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Joshua D Weatherston
- Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA
| | - Jeffrey D Cirillo
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Hung-Jen Wu
- Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
73
|
Mohammed M, Devnarain N, Elhassan E, Govender T. Exploring the applications of hyaluronic acid-based nanoparticles for diagnosis and treatment of bacterial infections. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1799. [PMID: 35485247 PMCID: PMC9539990 DOI: 10.1002/wnan.1799] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/22/2022] [Accepted: 03/24/2022] [Indexed: 12/12/2022]
Abstract
Hyaluronic acid (HA) has become a topic of significant interest in drug delivery research due to its excellent properties, including biosafety, biodegradability, and nonimmunogenicity. Moreover, due to its ease of modification, HA can be used to prepare several HA‐based nanosystems using various approaches. These approaches involve conjugating/grafting of hydrophobic moieties, polyelectrolytes complexation with cationic polymers, or surface modification of various nanoparticles using HA. These nanoparticles are able to selectively deliver antibacterial drugs or diagnostic molecules into the site of infections. In addition, HA can bind with overexpressed cluster of differentiation 44 (CD44) receptors in macrophages and also can be degraded by a family of enzymes called hyaluronidase (HAase) to release drugs or molecules. By binding with these receptors or being degraded at the infection site by HAase, HA‐based nanoparticles allow enhanced and targeted antibacterial delivery. Herein, we present a comprehensive and up‐to‐date review that highlights various techniques of preparation of HA‐based nanoparticles that have been reported in the literature. Furthermore, we also discuss and critically analyze numerous types of HA‐based nanoparticles that have been employed in antibacterial delivery to date. This article offers a critical overview of the potential of HA‐based nanoparticles to overcome the challenges of conventional antibiotics in the treatment of bacterial infections. Moreover, this review identifies further avenues of research for developing multifunctional and biomimetic HA‐based nanoparticles for the treatment, prevention, and/or detection of pathogenic bacteria. This article is categorized under:Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies
Collapse
Affiliation(s)
- Mahir Mohammed
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.,Faculty of Pharmacy, University of Khartoum, Khartoum, Sudan
| | - Nikita Devnarain
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Eman Elhassan
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Thirumala Govender
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
74
|
Decano JL, Iwamoto Y, Goto S, Lee JY, Matamalas JT, Halu A, Blaser M, Lee LH, Pieper B, Chelvanambi S, Silva-Nicolau J, Bartoli-Leonard F, Higashi H, Shibata H, Vyas P, Wang J, Gostjeva E, Body SC, Singh SA, Aikawa M, Aikawa E. A disease-driver population within interstitial cells of human calcific aortic valves identified via single-cell and proteomic profiling. Cell Rep 2022; 39:110685. [PMID: 35417712 DOI: 10.1016/j.celrep.2022.110685] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 08/04/2021] [Accepted: 03/24/2022] [Indexed: 11/03/2022] Open
Abstract
Cellular heterogeneity of aortic valves complicates the mechanistic evaluation of the calcification processes in calcific aortic valve disease (CAVD), and animal disease models are lacking. In this study, we identify a disease-driver population (DDP) within valvular interstitial cells (VICs). Through stepwise single-cell analysis, phenotype-guided omic profiling, and network-based analysis, we characterize the DDP fingerprint as CD44highCD29+CD59+CD73+CD45low and discover potential key regulators of human CAVD. These DDP-VICs demonstrate multi-lineage differentiation and osteogenic properties. Temporal proteomic profiling of DDP-VICs identifies potential targets for therapy, including MAOA and CTHRC1. In vitro loss-of-function experiments confirm our targets. Such a stepwise strategy may be advantageous for therapeutic target discovery in other disease contexts.
Collapse
Affiliation(s)
- Julius L Decano
- Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yukio Iwamoto
- Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Shinji Goto
- Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Janey Y Lee
- Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Joan T Matamalas
- Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Arda Halu
- Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mark Blaser
- Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lang Ho Lee
- Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Brett Pieper
- Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sarvesh Chelvanambi
- Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jessica Silva-Nicolau
- Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Francesca Bartoli-Leonard
- Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hideyuki Higashi
- Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Haruki Shibata
- Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Payal Vyas
- Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jianguo Wang
- Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Elena Gostjeva
- Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Simon C Body
- Boston University School of Medicine, Boston, MA 02118, USA
| | - Sasha A Singh
- Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Masanori Aikawa
- Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Cardiovascular Medicine, Center for Excellence in Vascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Elena Aikawa
- Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Cardiovascular Medicine, Center for Excellence in Vascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
75
|
ElSayed S, Jay GD, Cabezas R, Qadri M, Schmidt TA, Elsaid KA. Recombinant Human Proteoglycan 4 Regulates Phagocytic Activation of Monocytes and Reduces IL-1β Secretion by Urate Crystal Stimulated Gout PBMCs. Front Immunol 2022; 12:771677. [PMID: 34992596 PMCID: PMC8725049 DOI: 10.3389/fimmu.2021.771677] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/30/2021] [Indexed: 12/15/2022] Open
Abstract
Objectives To compare phagocytic activities of monocytes in peripheral blood mononuclear cells (PBMCs) from acute gout patients and normal subjects, examine monosodium urate monohydrate (MSU) crystal-induced IL-1β secretion ± recombinant human proteoglycan 4 (rhPRG4) or interleukin-1 receptor antagonist (IL-1RA), and study the anti-inflammatory mechanism of rhPRG4 in MSU stimulated monocytes. Methods Acute gout PBMCs were collected from patients in the Emergency Department and normal PBMCs were obtained from a commercial source. Monocytes in PBMCs were identified by flow cytometry. PBMCs were primed with Pam3CSK4 (1μg/mL) for 24h and phagocytic activation of monocytes was determined using fluorescently labeled latex beads. MSU (200μg/mL) stimulated IL-1β secretion was determined by ELISA. Reactive oxygen species (ROS) generation in monocytes was determined fluorometrically. PBMCs were incubated with IL-1RA (250ng/mL) or rhPRG4 (200μg/mL) and bead phagocytosis by monocytes was determined. THP-1 monocytes were treated with MSU crystals ± rhPRG4 and cellular levels of NLRP3 protein, pro-IL-1β, secreted IL-1β, and activities of caspase-1 and protein phosphatase-2A (PP2A) were quantified. The peritoneal influx of inflammatory and anti-inflammatory monocytes and neutrophils in Prg4 deficient mice was studied and the impact of rhPRG4 on immune cell trafficking was assessed. Results Enhanced phagocytic activation of gout monocytes under basal conditions (p<0.001) was associated with ROS generation and MSU stimulated IL-1β secretion (p<0.05). rhPRG4 reduced bead phagocytosis by normal and gout monocytes compared to IL-1RA and both treatments were efficacious in reducing IL-1β secretion (p<0.05). rhPRG4 reduced pro-IL-1β content, caspase-1 activity, conversion of pro-IL-1β to mature IL-1β and restored PP2A activity in monocytes (p<0.05). PP2A inhibition reversed rhPRG4’s effects on pro-IL-1β and mature IL-1β in MSU stimulated monocytes. Neutrophils accumulated in peritoneal cavities of Prg4 deficient mice (p<0.01) and rhPRG4 treatment reduced neutrophil accumulation and enhanced anti-inflammatory monocyte influx (p<0.05). Conclusions MSU phagocytosis was higher in gout monocytes resulting in higher ROS and IL-1β secretion. rhPRG4 reduced monocyte phagocytic activation to a greater extent than IL-1RA and reduced IL-1β secretion. The anti-inflammatory activity of rhPRG4 in monocytes is partially mediated by PP2A, and in vivo, PRG4 plays a role in regulating the trafficking of immune cells into the site of a gout flare.
Collapse
Affiliation(s)
- Sandy ElSayed
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, United States
| | - Gregory D Jay
- Department of Emergency Medicine, Rhode Island Hospital, Providence, RI, United States
| | - Ralph Cabezas
- Department of Emergency Medicine, Rhode Island Hospital, Providence, RI, United States
| | - Marwa Qadri
- Department of Pharmacology, School of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Tannin A Schmidt
- Biomedical Engineering Department, University of Connecticut Health Center, Farmington, CT, United States
| | - Khaled A Elsaid
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, United States
| |
Collapse
|
76
|
Choi S, Yoon M, Choi KY. Approaches for Regenerative Healing of Cutaneous Wound with an Emphasis on Strategies Activating the Wnt/β-Catenin Pathway. Adv Wound Care (New Rochelle) 2022; 11:70-86. [PMID: 33573472 PMCID: PMC9831250 DOI: 10.1089/wound.2020.1284] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Significance: In adult mammals, spontaneous repair of a cutaneous wound occurs slowly and leaves a scar with skin adnexa deficiencies. To accelerate cutaneous wound-healing rates and avoid scar formation, current studies have focused on regenerative therapies. Recent Advances: Emerging therapeutics for regenerative wound healing often focus on the use of growth factors and stem cells. However, these therapeutic approaches have limited routine clinical use due to high costs and technical requirements. Critical Issue: Understanding the molecular mechanisms involved in the signaling pathways for cutaneous wound healing and neogenic synthesis of the skin components is important for identification of novel targets for the development of regenerative wound-healing agents. Future Directions: The Wnt/β-catenin pathway is a well-known key player for enhancement of the overall healing process involving tissue regeneration via crosstalk with other signaling pathways. Strategies that activate the Wnt/β-catenin pathway via modulation of the pathway-controlling regulatory factors could provide effective therapeutic approaches for regenerative wound healing.
Collapse
Affiliation(s)
- Sehee Choi
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Minguen Yoon
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Kang-Yell Choi
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea.,CK Biotech, Inc., Seodaemun-Gu, Korea.,Correspondence: CK Biotech, Inc., Room 417, Engineering Research Park, 50 Yonsei Ro, Seodaemun-Gu 03722, Korea
| |
Collapse
|
77
|
Hussen J, Al-Sukruwah MA. The Impact of the Animal Housing System on Immune Cell Composition and Function in the Blood of Dromedary Camels. Animals (Basel) 2022; 12:ani12030317. [PMID: 35158641 PMCID: PMC8833619 DOI: 10.3390/ani12030317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/23/2022] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary The present study investigated the impacts of a change in animal housing system on selected parameters of the camel immune system. Samples collected from camels during a free-ranging time were compared with samples collected from the same camels during movement-restricted housing. Movement-restricted camels showed elevated myeloperoxidase activity in their serum, a significant shape-change of their neutrophils, and higher reactive oxygen species content in their monocytes and neutrophils. The leukogram pattern of the camels under restricted housing was characterized by increased numbers of neutrophils, eosinophils, lymphocytes, and monocytes. Within the lymphocyte population, only the helper T cells and B cells were expanded in animals under restricted housing. In addition, restricted housing modulated the expression of several cell surface antigens, including monocyte-polarization markers and cell adhesion molecules. Functional analysis of bacterial phagocytosis indicated impaired antibacterial function of phagocytes in camels under restricted housing. In summary, the present study identified significant changes in blood immune cell composition, phenotype, and function in dromedary camels under restricted-housing conditions, and suggests the development of an excitement leukogram in those animals. Abstract Background: The dromedary camel (Camelus dromedarius) is an important livestock animal of desert and semi-desert ecosystems. In recent years, several elements of the camel immune system have been characterized. Stress and excitement induced by animal housing represent the most important environmental factors with potential modulatory effects on the immune system. The present study evaluated the impacts of a restricted-housing system on some phenotypic and functional properties of blood leukocytes in dromedary camels. Methods: Immunofluorescence and flow cytometry were used to comparatively analyze samples collected from camels during a free-ranging time and samples collected from the same camels during movement-restricted housing. Results: In comparison to blood samples collected from the camels during the free-ranging time, samples from movement-restricted camels showed elevated serum myeloperoxidase activity, a significant shape-change in their neutrophils, and higher reactive oxygen species content in their monocytes and neutrophils, indicating increased cellular oxidative stress under movement-restricted housing. The leukogram pattern of the camels under restricted housing was characterized by leukocytosis with increased numbers of neutrophils, eosinophils, lymphocytes, and monocytes, resembling an excitement leukogram pattern. Within the lymphocyte population, only the helper T cells and B cells were expanded in animals under restricted housing. The upregulation of CD163 together with the downregulation of MHC-II on monocytes from excited camels indicate a modulatory potential of animal excitement to polarize monocytes toward an anti-inflammatory phenotype. Functional analysis of bacterial phagocytosis indicates an impaired antibacterial function of phagocytes in excited camels. The downregulation of several cell adhesion molecules on leukocytes from excited camels suggests a role for impaired cell adhesion and tissue migration and leukocyte retention in blood in the observed leukocytosis in animals under excitement. Conclusions: The present study identified significant changes in blood immune cell composition, phenotype, and function in dromedary camels under restricted-housing conditions. The observed changes in leukocyte composition suggest the development of an excitement leukogram pattern in camels under movement-restricted housing. To evaluate the clinical relevance of the observed changes in immune cell phenotype and function for the immune competence of camels under restricted housing, further studies are required.
Collapse
|
78
|
CD44-Targeted Carriers: The Role of Molecular Weight of Hyaluronic Acid in the Uptake of Hyaluronic Acid-Based Nanoparticles. Pharmaceuticals (Basel) 2022; 15:ph15010103. [PMID: 35056160 PMCID: PMC8781203 DOI: 10.3390/ph15010103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 02/01/2023] Open
Abstract
Nanotechnology offers advanced biomedical tools for diagnosis and drug delivery, stressing the value of investigating the mechanisms by which nanocarriers interact with the biological environment. Herein, the cellular response to CD44-targeted nanoparticles (NPs) was investigated. CD44, the main hyaluronic acid (HA) receptor, is widely exploited as a target for therapeutic purposes. HA NPs were produced by microfluidic platform starting from HA with different molecular weights (Mw, 280, 540, 820 kDa) by polyelectrolyte complexation with chitosan (CS). Thanks to microfluidic technology, HA/CS NPs with the same physical features were produced, and only the effects of HA Mw on CD44-overexpressing cells (human mesenchymal stem cells, hMSCs) were studied. This work provides evidence of the HA/CS NPs biocompatibility regardless the HA Mw and reveals the effect of low Mw HA in improving the cell proliferation. Special attention was paid to the endocytic mechanisms used by HA/CS NPs to enter hMSCs. The results show the notable role of CD44 and the pronounced effect of HA Mw in the NPs’ internalization. HA/CS NPs uptake occurs via different endocytic pathways simultaneously, and most notably, NPs with 280 kDa HA were internalized by clathrin-mediated endocytosis. Instead, NPs with 820 kDa HA revealed a greater contribution of caveolae and cytoskeleton components.
Collapse
|
79
|
Hsiao Y, Chi J, Li C, Chen L, Chen Y, Liang H, Lo Y, Hong J, Chuu C, Hung L, Du J, Chang W, Wang J. Disruption of the pentraxin 3/CD44 interaction as an efficient therapy for triple-negative breast cancers. Clin Transl Med 2022; 12:e724. [PMID: 35090088 PMCID: PMC8797470 DOI: 10.1002/ctm2.724] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/13/2022] [Accepted: 01/17/2022] [Indexed: 12/29/2022] Open
Abstract
Due to the heterogeneity and high frequency of genome mutations in cancer cells, targeting vital protumour factors found in stromal cells in the tumour microenvironment may represent an ideal strategy in cancer therapy. However, the regulation and mechanisms of potential targetable therapeutic candidates need to be investigated. An in vivo study demonstrated that loss of pentraxin 3 (PTX3) in stromal cells significantly decreased the metastasis and growth of cancer cells. Clinically, our results indicate that stromal PTX3 expression correlates with adverse prognostic features and is associated with worse survival outcomes in triple-negative breast cancer (TNBC). We also found that transforming growth factor beta 1 (TGF-β1) induces PTX3 expression by activating the transcription factor CCAAT/enhancer binding protein delta (CEBPD) in stromal fibroblasts. Following PTX3 stimulation, CD44, a PTX3 receptor, activates the downstream ERK1/2, AKT and NF-κB pathways to specifically contribute to the metastasis/invasion and stemness of TNBC MDA-MB-231 cells. Two types of PTX3 inhibitors were developed to disrupt the PTX3/CD44 interaction and they showed a significant effect on attenuating growth and restricting the metastasis/invasion of MDA-MB-231 cells, suggesting that targeting the PTX3/CD44 interaction could be a new strategy for future TNBC therapies.
Collapse
Affiliation(s)
- Yu‐Wei Hsiao
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Jhih‐Ying Chi
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Chien‐Feng Li
- Department of PathologyChi‐Mei Medical CenterTainanTaiwan R. O. C.
| | - Lei‐Yi Chen
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Yi‐Ting Chen
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Hsin‐Yin Liang
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Yu‐Chih Lo
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Jhen‐Yi Hong
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Chin‐Pin Chuu
- Institute of Cellular and System MedicineNational Health Research InstitutesMiaoli CountyTaiwan R. O. C.
| | - Liang‐Yi Hung
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Jyun‐Yi Du
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Wen‐Chang Chang
- Graduate Institute of Medical Sciences, College of MedicineTaipei Medical UniversityTaipeiTaiwan R. O. C.
| | - Ju‐Ming Wang
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
- Graduate Institute of Medical Sciences, College of MedicineTaipei Medical UniversityTaipeiTaiwan R. O. C.
- International Research Center for Wound Repair and RegenerationNational Cheng Kung UniversityTainanTaiwan R. O. C.
- Department of Physiology, College of MedicineNational Cheng Kung UniversityTainanTaiwan R. O. C.
- Graduate Institute of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan R. O. C.
| |
Collapse
|
80
|
Rao SS, Prabhu A, Kudkuli J, Surya S, Rekha P. Hyaluronic acid sustains platelet stability with prolonged growth factor release and accelerates wound healing by enhancing proliferation and collagen deposition in diabetic mice. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2021.102898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
81
|
Fopiano KA, Jalnapurkar S, Davila AC, Arora V, Bagi Z. Coronary Microvascular Dysfunction and Heart Failure with Preserved Ejection Fraction - implications for Chronic Inflammatory Mechanisms. Curr Cardiol Rev 2022; 18:e310821195986. [PMID: 34488616 PMCID: PMC9413735 DOI: 10.2174/1573403x17666210831144651] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 06/01/2021] [Accepted: 06/14/2021] [Indexed: 11/22/2022] Open
Abstract
Coronary Microvascular Dysfunction (CMD) is now considered one of the key underlying pathologies responsible for the development of both acute and chronic cardiac complications. It has been long recognized that CMD contributes to coronary no-reflow, which occurs as an acute complication during percutaneous coronary interventions. More recently, CMD was proposed to play a mechanistic role in the development of left ventricle diastolic dysfunction in heart failure with preserved ejection fraction (HFpEF). Emerging evidence indicates that a chronic low-grade pro-inflammatory activation predisposes patients to both acute and chronic cardiovascular complications raising the possibility that pro-inflammatory mediators serve as a mechanistic link in HFpEF. Few recent studies have evaluated the role of the hyaluronan-CD44 axis in inflammation-related cardiovascular pathologies, thus warranting further investigations. This review article summarizes current evidence for the role of CMD in the development of HFpEF, focusing on molecular mediators of chronic proinflammatory as well as oxidative stress mechanisms and possible therapeutic approaches to consider for treatment and prevention.
Collapse
Affiliation(s)
- Katie Anne Fopiano
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Sawan Jalnapurkar
- Division of Cardiology, Department of Medicine, Medical College of Georgia, Augusta University Augusta, GA 30912, USA
| | - Alec C Davila
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Vishal Arora
- Division of Cardiology, Department of Medicine, Medical College of Georgia, Augusta University Augusta, GA 30912, USA
| | - Zsolt Bagi
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
82
|
Immunogenetics of Lupus Erythematosus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1367:213-257. [DOI: 10.1007/978-3-030-92616-8_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
83
|
Corker A, Neff LS, Broughton P, Bradshaw AD, DeLeon-Pennell KY. Organized Chaos: Deciphering Immune Cell Heterogeneity's Role in Inflammation in the Heart. Biomolecules 2021; 12:11. [PMID: 35053159 PMCID: PMC8773626 DOI: 10.3390/biom12010011] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/10/2021] [Accepted: 12/18/2021] [Indexed: 12/24/2022] Open
Abstract
During homeostasis, immune cells perform daily housekeeping functions to maintain heart health by acting as sentinels for tissue damage and foreign particles. Resident immune cells compose 5% of the cellular population in healthy human ventricular tissue. In response to injury, there is an increase in inflammation within the heart due to the influx of immune cells. Some of the most common immune cells recruited to the heart are macrophages, dendritic cells, neutrophils, and T-cells. In this review, we will discuss what is known about cardiac immune cell heterogeneity during homeostasis, how these cell populations change in response to a pathology such as myocardial infarction or pressure overload, and what stimuli are regulating these processes. In addition, we will summarize technologies used to evaluate cell heterogeneity in models of cardiovascular disease.
Collapse
Affiliation(s)
- Alexa Corker
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC 29425, USA; (A.C.); (L.S.N.); (P.B.); (A.D.B.)
| | - Lily S. Neff
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC 29425, USA; (A.C.); (L.S.N.); (P.B.); (A.D.B.)
| | - Philip Broughton
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC 29425, USA; (A.C.); (L.S.N.); (P.B.); (A.D.B.)
| | - Amy D. Bradshaw
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC 29425, USA; (A.C.); (L.S.N.); (P.B.); (A.D.B.)
- Ralph H. Johnson Veterans Affairs Medical Center, Medical University of South Carolina, Charleston, SC 29401, USA
| | - Kristine Y. DeLeon-Pennell
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC 29425, USA; (A.C.); (L.S.N.); (P.B.); (A.D.B.)
- Ralph H. Johnson Veterans Affairs Medical Center, Medical University of South Carolina, Charleston, SC 29401, USA
| |
Collapse
|
84
|
Gholaminejad A, Roointan A, Gheisari Y. Transmembrane signaling molecules play a key role in the pathogenesis of IgA nephropathy: a weighted gene co-expression network analysis study. BMC Immunol 2021; 22:73. [PMID: 34861820 PMCID: PMC8642929 DOI: 10.1186/s12865-021-00468-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 11/19/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Immunoglobulin A nephropathy (IgAN) is one of the most common primary glomerulonephritis and a serious health concern worldwide; though still the underlying molecular mechanisms of IgAN are yet to be known and there is no efficient treatment for this disease. The main goal of this study was to explore the IgAN underlying pathogenic pathways, plus identifying the disease correlated modules and genes using the weighted gene co-expression network analysis (WGCNA) algorithm. RESULTS GSE104948 dataset (the expression data from glomerular tissue of IgAN patients) was analyzed and the identified differentially expressed genes (DEGs) were introduced to the WGCNA algorithm for building co-expression modules. Genes were classified into six co-expression modules. Genes of the disease's most correlated module were mainly enriched in the immune system, cell-cell communication and transmembrane cell signaling pathways. The PPI network was constructed by genes in all the modules and after hub-gene identification and validation steps, 11 genes, mostly transmembrane proteins (CD44, TLR1, TLR2, GNG11, CSF1R, TYROBP, ITGB2, PECAM1), as well as DNMT1, CYBB and PSMB9 were identified as potentially key players in the pathogenesis of IgAN. In the constructed regulatory network, hsa-miR-129-2-3p, hsa-miR-34a-5p and hsa-miR-27a-3p, as well as STAT3 were spotted as top molecules orchestrating the regulation of the hub genes. CONCLUSIONS The excavated hub genes from the hearts of co-expressed modules and the PPI network were mostly transmembrane signaling molecules. These genes and their upstream regulators could deepen our understanding of IgAN and be considered as potential targets for hindering its progression.
Collapse
Affiliation(s)
- Alieh Gholaminejad
- Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Hezar Jerib Avenue, 81746-73461, Isfahan, Iran
| | - Amir Roointan
- Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Hezar Jerib Avenue, 81746-73461, Isfahan, Iran.
| | - Yousof Gheisari
- Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Hezar Jerib Avenue, 81746-73461, Isfahan, Iran
| |
Collapse
|
85
|
Soliman F, Ye L, Jiang W, Hargest R. Targeting Hyaluronic Acid and Peritoneal Dissemination in Colorectal Cancer. Clin Colorectal Cancer 2021; 21:e126-e134. [PMID: 34955378 DOI: 10.1016/j.clcc.2021.11.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 10/30/2021] [Accepted: 11/22/2021] [Indexed: 11/03/2022]
Abstract
Peritoneal metastasis (PM) from colorectal cancer (CRC) carries a significant mortality rate for patients and treatment is challenging. The development of PM is a multistep process involving detachment, adhesion, invasion and colonization of the peritoneal cavity. Cytoreductive surgery and HIPEC (hyperthermic intraperitoneal chemotherapy) for PM from CRC has some benefit but overall survival is poor and recurrence rates are high. Treatments to prevent the development of peritoneal metastasis could have the potential to improve CRC survival and disease-free outcomes. The ability of cancer cells to invade the peritoneum and become established as metastatic tumors is influenced by a multifactorial process. Hyaluronic acid (HA) has been shown to coat the mesothelial cells of the peritoneum and has been demonstrated to be utilized in various malignancies as part of the metastatic process in peritoneal dissemination. CD44, RHAMM (CD168) and ICAM-1 have all been shown to be binding partners for HA. Targeting HA-mediated binding may prevent adhesion to distant sites within the peritoneum through suppression of interaction of these molecules. Here we review the current literature and discuss key molecules involved with PM dissemination, with the potential to target these mechanisms in the delivery of future treatments.
Collapse
Affiliation(s)
- Faris Soliman
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, School of Medicine, Cardiff University; Cardiff and Vale University Health Board.
| | - Lin Ye
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, School of Medicine, Cardiff University
| | - Wenguo Jiang
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, School of Medicine, Cardiff University
| | - Rachel Hargest
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, School of Medicine, Cardiff University; Cardiff and Vale University Health Board
| |
Collapse
|
86
|
Terracciano R, Carcamo-Bahena Y, Butler EB, Demarchi D, Grattoni A, Filgueira CS. Hyaluronate-Thiol Passivation Enhances Gold Nanoparticle Peritumoral Distribution When Administered Intratumorally in Lung Cancer. Biomedicines 2021; 9:1561. [PMID: 34829790 PMCID: PMC8615404 DOI: 10.3390/biomedicines9111561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/21/2021] [Accepted: 10/23/2021] [Indexed: 12/13/2022] Open
Abstract
Biofouling is the unwanted adsorption of cells, proteins, or intracellular and extracellular biomolecules that can spontaneously occur on the surface of metal nanocomplexes. It represents a major issue in bioinorganic chemistry because it leads to the creation of a protein corona, which can destabilize a colloidal solution and result in undesired macrophage-driven clearance, consequently causing failed delivery of a targeted drug cargo. Hyaluronic acid (HA) is a bioactive, natural mucopolysaccharide with excellent antifouling properties, arising from its hydrophilic and polyanionic characteristics in physiological environments which prevent opsonization. In this study, hyaluronate-thiol (HA-SH) (MW 10 kDa) was used to surface-passivate gold nanoparticles (GNPs) synthesized using a citrate reduction method. HA functionalized GNP complexes (HA-GNPs) were characterized using absorption spectroscopy, scanning electron microscopy, zeta potential, and dynamic light scattering. GNP cellular uptake and potential dose-dependent cytotoxic effects due to treatment were evaluated in vitro in HeLa cells using inductively coupled plasma-optical emission spectrometry (ICP-OES) and trypan blue and MTT assays. Further, we quantified the in vivo biodistribution of intratumorally injected HA functionalized GNPs in Lewis Lung carcinoma (LLC) solid tumors grown on the flank of C57BL/6 mice and compared localization and retention with nascent particles. Our results reveal that HA-GNPs show overall greater peritumoral distribution (** p < 0.005, 3 days post-intratumoral injection) than citrate-GNPs with reduced biodistribution in off-target organs. This property represents an advantageous step forward in localized delivery of metal nano-complexes to the infiltrative region of a tumor, which may improve the application of nanomedicine in the diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Rossana Terracciano
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (R.T.); (Y.C.-B.); (A.G.)
- Department of Electronics and Telecommunications, Politecnico di Torino, 10129 Torino, Italy;
| | - Yareli Carcamo-Bahena
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (R.T.); (Y.C.-B.); (A.G.)
| | - E. Brian Butler
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA;
| | - Danilo Demarchi
- Department of Electronics and Telecommunications, Politecnico di Torino, 10129 Torino, Italy;
| | - Alessandro Grattoni
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (R.T.); (Y.C.-B.); (A.G.)
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA;
- Department of Surgery, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Carly S. Filgueira
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (R.T.); (Y.C.-B.); (A.G.)
- Department of Cardiovascular Surgery, Houston Methodist Research Institute, Houston, TX 77030, USA
| |
Collapse
|
87
|
Chiesa E, Greco A, Riva F, Dorati R, Conti B, Modena T, Genta I. Hyaluronic Acid-Based Nanoparticles for Protein Delivery: Systematic Examination of Microfluidic Production Conditions. Pharmaceutics 2021; 13:1565. [PMID: 34683858 PMCID: PMC8539066 DOI: 10.3390/pharmaceutics13101565] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/09/2021] [Accepted: 09/23/2021] [Indexed: 11/17/2022] Open
Abstract
Hyaluronic acid-based nanoparticles (HA NPs) can be used to deliver a protein cargo to cells overexpressing HA receptors such as CD44 since they combine the low toxicity of the carrier and the retention of the protein integrity with the receptor-mediated internalization. HA properties play a crucial but sometimes unclear role in managing the formation and stability of the meshwork, cell interactions, and ultimately the protein entrapment efficacy. Nowadays, microfluidic is an innovative technology that allows to overcome limits linked to the NPs production, guaranteeing reproducibility and control of individual batches. Taking advantage of this technique, in this research work, the role of HA weight average molecular weight (Mw) in NPs formation inside a microfluidic device has been specifically faced. Based on the relationship between polymer Mw and solution viscosity, a methodological approach has been proposed to ensure critical quality attributes (size of 200 nm, PDI ≤ 0.3) to NPs made by HA with different Mw (280, 540, 710 and 820 kDa). The feasibility of the protein encapsulation was demonstrated by using Myoglobin, as a model neutral protein, with an encapsulation efficiency always higher than 50%. Lastly, all NPs samples were successfully internalized by CD44-expressing cells.
Collapse
Affiliation(s)
- Enrica Chiesa
- Department of Surgery, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Antonietta Greco
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (A.G.); (R.D.); (B.C.); (T.M.)
| | - Federica Riva
- Department of Public Health, Experimental and Forensic Medicine, Histology and Embryology Unit, University of Pavia, 27100 Pavia, Italy;
| | - Rossella Dorati
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (A.G.); (R.D.); (B.C.); (T.M.)
| | - Bice Conti
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (A.G.); (R.D.); (B.C.); (T.M.)
| | - Tiziana Modena
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (A.G.); (R.D.); (B.C.); (T.M.)
| | - Ida Genta
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (A.G.); (R.D.); (B.C.); (T.M.)
| |
Collapse
|
88
|
Hundhausen C, Schneckmann R, Ostendorf Y, Rimpler J, von Glinski A, Kohlmorgen C, Pasch N, Rolauer L, von Ameln F, Eckermann O, Altschmied J, Ale-Agha N, Haendeler J, Flögel U, Fischer JW, Grandoch M. Endothelial hyaluronan synthase 3 aggravates acute colitis in an experimental model of inflammatory bowel disease. Matrix Biol 2021; 102:20-36. [PMID: 34464693 DOI: 10.1016/j.matbio.2021.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/27/2021] [Accepted: 08/23/2021] [Indexed: 02/08/2023]
Abstract
The association between hyaluronan (HA) accumulation and increased inflammation in the colon suggests that HA is a potential therapeutic target in inflammatory bowel disease (IBD). However, whether patients with IBD would benefit from interference with HA synthesis is unknown. Here, we used pharmacological and genetic approaches to investigate the impact of systemic and partial blockade of HA synthesis in the Dextran Sodium Sulfate (DSS)-induced colitis model. To systemically inhibit HA production, we used 4-Methylumbelliferone (4-MU), whereas genetic approaches included the generation of mice with global or inducible cell-type specific deficiency in the Hyaluronan synthase 3 (Has3). We found that 4-MU treatment did not ameliorate but exacerbated disease severity characterized by increased body weight loss and enhanced colon tissue destruction compared to control mice without colitis. In contrast, global Has3 deficiency had a profound protective effect as reflected by a low colitis score and reduced infiltration of immune cells into the colon. To get further mechanistic insight into the proinflammatory role of HAS3, we deleted Has3 in a cell-type specific manner. Interestingly, while lack of Has3 expression in intestinal epithelial and smooth muscle cells had no effect or was rather proinflammatory, mice with Has3 deficiency in the endothelium were strongly protected against acute colitis. We conclude that endothelium-derived HAS3 plays a critical role in driving experimental colitis, warranting future studies on cell type-specific therapeutic interference with HA production in human IBD.
Collapse
Affiliation(s)
- Christian Hundhausen
- Institute for Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Rebekka Schneckmann
- Institute for Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Yanina Ostendorf
- Institute for Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Jacqueline Rimpler
- Institute for Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Anette von Glinski
- Institute for Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Christina Kohlmorgen
- Institute for Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Nina Pasch
- Institute for Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Luca Rolauer
- Institute for Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Florian von Ameln
- Environmentally-induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Clinics and Heinrich-Heine-University Düsseldorf and IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Olaf Eckermann
- Environmentally-induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Clinics and Heinrich-Heine-University Düsseldorf and IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Joachim Altschmied
- Environmentally-induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Clinics and Heinrich-Heine-University Düsseldorf and IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Niloofar Ale-Agha
- Environmentally-induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Clinics and Heinrich-Heine-University Düsseldorf, Germany
| | - Judith Haendeler
- Environmentally-induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Clinics and Heinrich-Heine-University Düsseldorf, Germany
| | - Ulrich Flögel
- Institute for Molecular Cardiology, University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Jens W Fischer
- Institute for Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Maria Grandoch
- Institute for Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University Düsseldorf, Germany.
| |
Collapse
|
89
|
Pirri C, Fede C, Pirri N, Petrelli L, Fan C, De Caro R, Stecco C. Diabetic Foot: The Role of Fasciae, a Narrative Review. BIOLOGY 2021; 10:biology10080759. [PMID: 34439991 PMCID: PMC8389550 DOI: 10.3390/biology10080759] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/15/2021] [Accepted: 08/04/2021] [Indexed: 12/19/2022]
Abstract
Simple Summary Diabetes mellitus and its complications are increasingly prevalent worldwide with severe impacts on patients and health care systems. Diabetic foot ulcers have an important impact on disability, morbidity, and mortality. The mechanism of diabetic wound chronicity has not yet been understood in a complete way. Regarding the involved soft tissues, little space has been given to the fasciae, even if nowadays there is more and more evidence of their role in proprioception, muscular force transmission, skin vascularization and tropism, and wound healing. Thus, we aimed to deepen the fascial involvement in diabetic wounds. Based on this review, we suggest that a clear scientific perception of fascial role can improve treatment strategies and create new perspectives of treatment. Abstract Wound healing is an intricate, dynamic process, in which various elements such as hyperglycemia, neuropathy, blood supply, matrix turnover, wound contraction, and the microbiome all have a role in this “out of tune” diabetic complex symphony, particularly noticeable in the complications of diabetic foot. Recently it was demonstrated that the fasciae have a crucial role in proprioception, muscular force transmission, skin vascularization and tropism, and wound healing. Indeed, the fasciae are a dynamic multifaceted meshwork of connective tissue comprised of diverse cells settled down in the extracellular matrix and nervous fibers; each constituent plays a particular role in the fasciae adapting in various ways to the diverse stimuli. This review intends to deepen the discussion on the possible fascial role in diabetic wounds. In diabetes, the thickening of collagen, the fragmentation of elastic fibers, and the changes in glycosaminoglycans, in particular hyaluronan, leads to changes in the stiffness, gliding, and the distribution of force transmission in the fasciae, with cascading repercussions at the cellular and molecular levels, consequently feeding a vicious pathophysiological circle. A clear scientific perception of fascial role from microscopic and macroscopic points of view can facilitate the identification of appropriate treatment strategies for wounds in diabetes and create new perspectives of treatment.
Collapse
Affiliation(s)
- Carmelo Pirri
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, 35121 Padua, Italy; (C.F.); (L.P.); (C.F.); (R.D.C.); (C.S.)
- Correspondence:
| | - Caterina Fede
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, 35121 Padua, Italy; (C.F.); (L.P.); (C.F.); (R.D.C.); (C.S.)
| | - Nina Pirri
- School of Medicine and Surgery, University of Messina, 98125 Messina, Italy;
| | - Lucia Petrelli
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, 35121 Padua, Italy; (C.F.); (L.P.); (C.F.); (R.D.C.); (C.S.)
| | - Chenglei Fan
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, 35121 Padua, Italy; (C.F.); (L.P.); (C.F.); (R.D.C.); (C.S.)
| | - Raffaele De Caro
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, 35121 Padua, Italy; (C.F.); (L.P.); (C.F.); (R.D.C.); (C.S.)
| | - Carla Stecco
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, 35121 Padua, Italy; (C.F.); (L.P.); (C.F.); (R.D.C.); (C.S.)
| |
Collapse
|
90
|
Human Umbilical Cord Mesenchymal Stem Cells in Combination with Hyaluronic Acid Ameliorate the Progression of Knee Osteoarthritis. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11146650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The aim of this study is to evaluate the feasibility and usefulness of the human umbilical cord mesenchymal stem cells (hUC-MSCs) and hyaluronan acid (HA) combination to attenuate osteoarthritis progression in the knee while simultaneously providing some insights on the mitigation mechanism. In vitro, the effect of hUC-MSCs with HA treatment on chondrocyte cell viability and the cytokine profile were analyzed. Additionally, the antioxidation capability of hUC-MSCs-CM (conditioned medium) with HA towards H2O2-induced chondrocyte cell damage was evaluated. The HA addition increased the hUC-MSC antioxidation capability and cytokine secretion, such as Dickkopf-related protein 1 (DKK-1) and hepatocyte growth factor (HGF), while no adverse effect on the cell viability was observed. In vivo, the intra-articular injection of hUC-MSCs with HA to a mono-iodoacetate (MIA)-induced knee osteoarthritis (KOA) rat model was performed and investigated. Attenuation of the KOA progression in the MIA-damaged rat model was seen best in hUC-MSCs with a HA combination compared to the vehicle control or each individual element. Combining hUC-MSCs and HA resulted in a synergistic effect, such as increasing the cell therapeutic capability while incurring no observable adverse effects. Therefore, this combinatorial therapy is feasible and has promising potential to ameliorate KOA progression.
Collapse
|
91
|
Krešić N, Prišlin M, Vlahović D, Kostešić P, Ljolje I, Brnić D, Turk N, Musulin A, Habrun B. The Expression Pattern of Surface Markers in Canine Adipose-Derived Mesenchymal Stem Cells. Int J Mol Sci 2021; 22:ijms22147476. [PMID: 34299095 PMCID: PMC8303761 DOI: 10.3390/ijms22147476] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/30/2021] [Accepted: 07/09/2021] [Indexed: 11/16/2022] Open
Abstract
The influence of cultivation on the expression pattern of canine adipose-derived mesenchymal stem cells (cAD-MSCs) surface markers, contributing to, among others, the promotion of growth, proliferation, differentiation and immunomodulatory mechanisms of an excellent therapeutic, is still unknown. To fill the gap, we investigated CD90, CD44, CD73, CD29, CD271, CD105, CD45 and CD14 patterns of expression at the protein level with flow cytometry and mRNA level using a real-time polymerase chain reaction array. Gentle variations of expression occurred during cultivation, along with increased CD90, CD44 and CD29 expression, low and decreasing CD271 and CD73 expression and a decrease of initially high CD105. As expected, CD45 and CD14 were not expressed by cAD-MSCs. Interestingly, we discovered a significant decrease of CD73 expression, compared to early (P1–P3) to late (P4–P6) passages, although the CD73 gene expression was found to be stable. The percentage of positive cells was found to be higher for all positive markers up to P4. As CD73′s one important feature is a modulation from a pro-inflammatory environment to an anti-inflammatory milieu, the expression of CD73 in our conditions indicate the need to consider the time cells spend in vitro before being transplanted into patients, since it could impact their favourable therapeutical properties.
Collapse
Affiliation(s)
- Nina Krešić
- Virology Department, Croatian Veterinary Institute, Savska Cesta 143, 10 000 Zagreb, Croatia; (M.P.); (D.B.)
- Correspondence:
| | - Marina Prišlin
- Virology Department, Croatian Veterinary Institute, Savska Cesta 143, 10 000 Zagreb, Croatia; (M.P.); (D.B.)
| | - Dunja Vlahović
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10 000 Zagreb, Croatia;
| | - Petar Kostešić
- Surgery, Orthopaedics and Ophthalmology Clinic, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10 000 Zagreb, Croatia; (P.K.); (A.M.)
| | - Ivana Ljolje
- Veterinary Clinic for Small Animals Buba, Dore Pfanove 11, 10 000 Zagreb, Croatia;
| | - Dragan Brnić
- Virology Department, Croatian Veterinary Institute, Savska Cesta 143, 10 000 Zagreb, Croatia; (M.P.); (D.B.)
| | - Nenad Turk
- Department of Microbiology and Infectious Diseases with Clinic, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10 000 Zagreb, Croatia;
| | - Andrija Musulin
- Surgery, Orthopaedics and Ophthalmology Clinic, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10 000 Zagreb, Croatia; (P.K.); (A.M.)
| | - Boris Habrun
- Department for Bacteriology and Parasitology, Croatian Veterinary Institute, Savska Cesta 143, 10 000 Zagreb, Croatia;
| |
Collapse
|
92
|
Michon A. Hyaluronic acid soft tissue filler delayed inflammatory reaction following COVID-19 vaccination - A case report. J Cosmet Dermatol 2021; 20:2684-2690. [PMID: 34174156 PMCID: PMC8447415 DOI: 10.1111/jocd.14312] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/02/2021] [Accepted: 06/16/2021] [Indexed: 12/02/2022]
Abstract
Background The use of hyaluronic acid soft tissue fillers in aesthetic medicine exploded in recent years for many reasons, including being relatively safe. Incidence of delayed inflammatory reactions (DIRs) to hyaluronic acid soft tissue fillers range between 0.3% and 4.25%. These reactions are mediated by T‐lymphocytes and can be triggered by flu‐like illnesses, including SARS‐CoV‐2 infection. Vaccination may also induce hypersensitivity. Aim In this case report, we present two cases of delayed reaction after hyaluronic acid soft tissue filler treatment of the tear trough area and following mRNA vaccination against SARS‐Cov‐2, also known as COVID‐19, months later. Patients A 39‐year old female who previously had her tear trough area treated with hyaluronic acid soft tissue filler developed swelling days after getting the mRNA Pfizer‐BioNTech COVID‐19 vaccine. Another patient, a 61‐year‐olf female, developed intermittent facial swelling in areas previously treated with hyaluronic acid soft tissue fillers days after receiving her first dose of the mRNA Pfizer‐BioNTech COVID‐19 vaccine. Results As demonstrated in our case report, vaccination against COVID‐19 may induce DIRs in patients who previously had hyaluronic soft tissue fillers. Conclusion Delayed inflammatory reactions to hyaluronic acid soft tissue fillers are uncommon and usually self‐limited, with frequent spontaneous resolution. However, considering the ongoing pandemic and the worldwide demand for vaccines against COVID‐19, the aesthetic providers should be conscious of the risks posed by the interaction of such vaccines in patients who previously had or seeking hyaluronic acid soft tissue filler injections.
Collapse
Affiliation(s)
- Alain Michon
- The Ottawa Skin Clinic, Ottawa, ON, Canada.,Institut du Savoir Montfort, Montfort Hospital, Ottawa, ON, Canada.,Department of Emergency Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
93
|
Charan TR, Bhutto MA, Bhutto MA, Tunio AA, Khuhro GM, Khaskheli SA, Mughal AA. “Nanomaterials of curcumin-hyaluronic acid”: their various methods of formulations, clinical and therapeutic applications, present gap, and future directions. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2021. [DOI: 10.1186/s43094-021-00281-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Abstract
Background
Nanomaterials of curcumin with hyaluronic acid have gained a lot of attention for potential therapeutic applications of curcumin and hyaluronic acid with or without other additional drugs. Overall studies of curcumin and hyaluronic acid show that nanomaterials of curcumin with hyaluronic acid accelerate the efficacy of curcumin in the treatment of various disorders like arthritis, cancer, hepatic fibrosis, neural disorders, wound healing, and skin regeneration, it is largely due to the combined effect of hyaluronic acid and curcumin. However, due to limited clinical trials and experiments on humans and animals, there is a substantial gap in research for the safety and efficacy of nanomaterials of curcumin-hyaluronic acid in the treatment of curcumin and hyaluronic acid targeted diseases and disorders.
Main body of the abstract
In this current review, we have first described various reported synthetic nanomaterials of curcumin-hyaluronic acid, then in the next section, we have described various fields, disorders, and diseases where these are being applied and in the final section of this review, we discussed the research gap, and future research directions needed to propose the fabricated nanocurcumin-hyaluronic acid biomaterials.
Short conclusion
There are substantial gaps in research for the safety and efficacy of nanomaterials of curcumin with hyaluronic acid due to limited available data of clinical trials and experiments of nanocurcumin-hyaluronic acid biomaterials on humans and animals. So, it entirely requires serious and committed efforts through the well-organized system of practical and clinical trials which provide results, data, and detections that lead to the formulation of the best drug from curcumin with hyaluronic acid for the treatment of curcumin and hyaluronic acid targeted diseases and disorders.
Collapse
|
94
|
Mahajan K, Rojekar S, Desai D, Kulkarni S, Bapat G, Zinjarde S, Vavia P. Layer-by-Layer Assembled Nanostructured Lipid Carriers for CD-44 Receptor-Based Targeting in HIV-Infected Macrophages for Efficient HIV-1 Inhibition. AAPS PharmSciTech 2021; 22:171. [PMID: 34100170 DOI: 10.1208/s12249-021-01981-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 03/04/2021] [Indexed: 02/01/2023] Open
Abstract
Macrophages act as a cellular reservoir in HIV infection. Elimination of HIV from macrophages has been an unfulfilled dream due to the failure of drugs to reach them. To address this, we developed CD44 receptor-targeted, novel hyaluronic acid (HA)-coated nanostructured lipid carriers (NLCs) of efavirenz via washless layer-by-layer (LbL) assembly of HA and polyallylamine hydrochloride (PAH). NLCs were subjected to TEM analysis, size and zeta potential, in vitro release and encapsulation efficiency studies. The uptake of NLCs in THP-1 cells was studied using fluorescence microscopy and flow cytometry. The anti-HIV efficacy was evaluated using p24 antigen inhibition assay. NLCs were found to be spherical in shape with anionic zeta potential (-23.66 ± 0.87 mV) and 241.83 ± 5.38 nm particle size. NLCs exhibited prolonged release of efavirenz during in vitro drug release studies. Flow cytometry revealed 1.73-fold higher uptake of HA-coated NLCs in THP-1 cells. Cytotoxicity studies showed no significant change in cell viability in presence of NLCs as compared with the control. HA-coated NLCs distributed throughout the cell including cytoplasm, plasma membrane and nucleus, as observed during fluorescence microscopy. HA-coated NLCs demonstrated consistent and significantly higher inhibition (81.26 ± 1.70%) of p24 antigen which was 2.08-fold higher than plain NLCs. The obtained results suggested preferential uptake of HA-coated NLCs via CD44-mediated uptake. The present finding demonstrates that HA-based CD44 receptor targeting in HIV infection is an attractive strategy for maximising the drug delivery to macrophages and achieve effective viral inhibition.
Collapse
|
95
|
The pleiotropic effects of Prunus avium L. extract against oxidative stress on human fibroblasts. An in vitro approach. Mol Biol Rep 2021; 48:4441-4448. [PMID: 34100152 DOI: 10.1007/s11033-021-06464-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 06/01/2021] [Indexed: 10/24/2022]
Abstract
There is a persistent interest in innovative and multifunctional ingredients in biology research. With regards to this, natural sources have an important role due to their multiple benefits. Thus, this study aims to present the pleiotropic activity of Prunus avium L. extract on human primary fibroblasts for proving its efficacy in dermis-related processes. We focused on the safety and efficacy assessments based on cytotoxicity and gene expression analysis under oxidative stress. Specifically, Prunus avium L. extract was proved non-cytotoxic in human fibroblasts. The gene expression analysis unveiled that this extract has in vitro protective properties on human dermal fibroblasts under oxidative stress related to antioxidant activity, anti-inflammatory response, cell proliferation and cell- aging. Our study demonstrated for the very first time that the Prunus avium L. extract is a multifunctional ingredient as it mediates several human dermis-related in vitro processes highlighting its potential to be used as an active ingredient in skin care products.
Collapse
|
96
|
Kim H, Woo J, Dan K, Lee KM, Jin MS, Park IA, Ryu HS, Han D. Quantitative Proteomics Reveals Knockdown of CD44 Promotes Proliferation and Migration in Claudin-Low MDA-MB-231 and Hs 578T Breast Cancer Cell Lines. J Proteome Res 2021; 20:3720-3733. [PMID: 34075748 DOI: 10.1021/acs.jproteome.1c00293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CD44 is a transmembrane glycoprotein that can regulate the oncogenic process. This is known to be a marker of the claudin-low subtype of breast cancer, as well as a cancer stem cell marker. However, its functional regulatory roles are poorly understood in claudin-low breast cancer. To gain comprehensive insight into the function of CD44, we performed an in-depth tandem mass tag-based proteomic analysis of two claudin-low breast cancer cell lines (MDA-MB-231 and Hs 578T) transfected with CD44 siRNA. As a result, we observed that 2736 proteins were upregulated and 2172 proteins were downregulated in CD44-knockdown MDA-MB-231 cells. For Hs 578T CD44-knockdown cells, 412 proteins were upregulated and 443 were downregulated. Gene ontology and network analyses demonstrated that the suppression of this marker mediates significant functional alterations related to oncogenic cellular processes, including proliferation, metabolism, adhesion, and gene expression regulation. A functional study confirmed that CD44 knockdown inhibited proliferation by regulating the expression of genes related to cell cycle, translation, and transcription. Moreover, this promoted the expression of multiple cell adhesion-associated proteins and attenuated cancer cell migration. Finally, our proteomic study defines the landscape of the CD44-regulated proteome of claudin-low breast cancer cells, revealing changes that mediate cell proliferation and migration. Our proteomics data set has been deposited to the ProteomeXchange Consortium via the PRIDE repository with the data set identifier PXD015171.
Collapse
Affiliation(s)
- Hyeyoon Kim
- Department of Pathology, Seoul National University Hospital, Seoul 03080, Korea.,Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Korea.,Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul 03082, Korea
| | - Jongmin Woo
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99354, United States
| | - Kisoon Dan
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul 03082, Korea
| | - Kyung-Min Lee
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea
| | - Min-Sun Jin
- Department of Pathology, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Bucheon, Gyeonggi-do 14647, Korea
| | - In Ae Park
- Department of Pathology, Seoul National University Hospital, Seoul 03080, Korea.,Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Han Suk Ryu
- Department of Pathology, Seoul National University Hospital, Seoul 03080, Korea.,Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Korea.,Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea
| | - Dohyun Han
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul 03082, Korea
| |
Collapse
|
97
|
Rebendenne A, Roy P, Bonaventure B, Chaves VAL, Desmarets L, Rouillé Y, Tauziet M, Arnaud-Arnould M, Giovannini D, Lee Y, DeWeirdt P, Hegde M, Garcia de GF, McKellar J, Wencker M, Dubuisson J, Belouzard S, Moncorgé O, Doench JG, Goujon C. Bidirectional genome-wide CRISPR screens reveal host factors regulating SARS-CoV-2, MERS-CoV and seasonal HCoVs. RESEARCH SQUARE 2021:rs.3.rs-555275. [PMID: 34075371 PMCID: PMC8168385 DOI: 10.21203/rs.3.rs-555275/v1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Several genome-wide CRISPR knockout screens have been conducted to identify host factors regulating SARS-CoV-2 replication, but the models used have often relied on overexpression of ACE2 receptor. Additionally, such screens have yet to identify the protease TMPRSS2, known to be important for viral entry at the plasma membrane. Here, we conducted a meta-analysis of these screens and showed a high level of cell-type specificity of the identified hits, arguing for the necessity of additional models to uncover the full landscape of SARS-CoV-2 host factors. We performed genome-wide knockout and activation CRISPR screens in Calu-3 lung epithelial cells, as well as knockout screens in Caco-2 intestinal cells. In addition to identifying ACE2 and TMPRSS2 as top hits, our study reveals a series of so far unidentified and critical host-dependency factors, including the Adaptins AP1G1 and AP1B1 and the flippase ATP8B1. Moreover, new anti-SARS-CoV-2 proteins with potent activity, including several membrane-associated Mucins, IL6R, and CD44 were identified. We further observed that these genes mostly acted at the critical step of viral entry, with the notable exception of ATP8B1, the knockout of which prevented late stages of viral replication. Exploring the pro- and anti-viral breadth of these genes using highly pathogenic MERS-CoV, seasonal HCoV-NL63 and -229E and influenza A orthomyxovirus, we reveal that some genes such as AP1G1 and ATP8B1 are general coronavirus cofactors. In contrast, Mucins recapitulated their known role as a general antiviral defense mechanism. These results demonstrate the value of considering multiple cell models and perturbational modalities for understanding SARS-CoV-2 replication and provide a list of potential new targets for therapeutic interventions.
Collapse
Affiliation(s)
| | - Priyanka Roy
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, USA
| | | | | | - Lowiese Desmarets
- Lille University, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, France
| | - Yves Rouillé
- Lille University, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, France
| | | | | | | | - Yenarae Lee
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, USA
| | - Peter DeWeirdt
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, USA
| | - Mudra Hegde
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, USA
| | | | | | | | - Jean Dubuisson
- Lille University, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, France
| | - Sandrine Belouzard
- Lille University, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, France
| | | | - John G. Doench
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, USA
| | | |
Collapse
|
98
|
Nivedita PS, Shettar AK, Joy HH. Applications of Polysaccharides in Nutrition and Medicine. POLYSACCHARIDES 2021. [DOI: 10.1002/9781119711414.ch30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
99
|
Rebendenne A, Roy P, Bonaventure B, Chaves Valadão AL, Desmarets L, Rouillé Y, Tauziet M, Arnaud-Arnould M, Giovannini D, Lee Y, DeWeirdt P, Hegde M, Garcia de Gracia F, McKellar J, Wencker M, Dubuisson J, Belouzard S, Moncorgé O, Doench JG, Goujon C. Bidirectional genome-wide CRISPR screens reveal host factors regulating SARS-CoV-2, MERS-CoV and seasonal coronaviruses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 34031654 DOI: 10.1101/2021.05.19.444823] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Several genome-wide CRISPR knockout screens have been conducted to identify host factors regulating SARS-CoV-2 replication, but the models used have often relied on overexpression of ACE2 receptor. Additionally, such screens have yet to identify the protease TMPRSS2, known to be important for viral entry at the plasma membrane. Here, we conducted a meta-analysis of these screens and showed a high level of cell-type specificity of the identified hits, arguing for the necessity of additional models to uncover the full landscape of SARS-CoV-2 host factors. We performed genome-wide knockout and activation CRISPR screens in Calu-3 lung epithelial cells, as well as knockout screens in Caco-2 intestinal cells. In addition to identifying ACE2 and TMPRSS2 as top hits, our study reveals a series of so far unidentified and critical host-dependency factors, including the Adaptins AP1G1 and AP1B1 and the flippase ATP8B1. Moreover, new anti-SARS-CoV-2 proteins with potent activity, including several membrane-associated Mucins, IL6R, and CD44 were identified. We further observed that these genes mostly acted at the critical step of viral entry, with the notable exception of ATP8B1, the knockout of which prevented late stages of viral replication. Exploring the pro- and anti-viral breadth of these genes using highly pathogenic MERS-CoV, seasonal HCoV-NL63 and -229E and influenza A orthomyxovirus, we reveal that some genes such as AP1G1 and ATP8B1 are general coronavirus cofactors. In contrast, Mucins recapitulated their known role as a general antiviral defense mechanism. These results demonstrate the value of considering multiple cell models and perturbational modalities for understanding SARS-CoV-2 replication and provide a list of potential new targets for therapeutic interventions.
Collapse
|
100
|
Zhou L, Hao Q, Sugita S, Naito Y, He H, Yeh CC, Lee JW. Role of CD44 in increasing the potency of mesenchymal stem cell extracellular vesicles by hyaluronic acid in severe pneumonia. Stem Cell Res Ther 2021; 12:293. [PMID: 34016170 PMCID: PMC8136222 DOI: 10.1186/s13287-021-02329-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 04/07/2021] [Indexed: 12/17/2022] Open
Abstract
Background Although promising, clinical translation of human mesenchymal stem or stromal cell-derived extracellular vesicles (MSC EV) for acute lung injury is potentially limited by significant production costs. The current study was performed to determine whether pretreatment of MSC EV with high molecular weight hyaluronic acid (HMW HA) would increase the therapeutic potency of MSC EV in severe bacterial pneumonia. Methods In vitro experiments were performed to determine the binding affinity of HMW HA to MSC EV and its uptake by human monocytes, and whether HMW HA primed MSC EV would increase bacterial phagocytosis by the monocytes. In addition, the role of CD44 receptor on MSC EV in the therapeutic effects of HMW HA primed MSC EV were investigated. In Pseudomonas aeruginosa (PA) pneumonia in mice, MSC EV primed with or without HMW HA were instilled intravenously 4 h after injury. After 24 h, the bronchoalveolar lavage fluid, blood, and lungs were analyzed for levels of bacteria, inflammation, MSC EV trafficking, and lung pathology. Results MSC EV bound preferentially to HMW HA at a molecular weight of 1.0 MDa compared with HA with a molecular weight of 40 KDa or 1.5 MDa. HMW HA primed MSC EV further increased MSC EV uptake and bacterial phagocytosis by monocytes compared to treatment with MSC EV alone. In PA pneumonia in mice, instillation of HMW HA primed MSC EV further reduced inflammation and decreased the bacterial load by enhancing the trafficking of MSC EV to the injured alveolus. CD44 siRNA pretreatment of MSC EV prior to incubation with HMW HA eliminated its trafficking to the alveolus and therapeutic effects. Conclusions HMW HA primed MSC EV significantly increased the potency of MSC EV in PA pneumonia in part by enhancing the trafficking of MSC EV to the sites of inflammation via the CD44 receptor on MSC EV which was associated with increased antimicrobial activity. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02329-2.
Collapse
Affiliation(s)
- Li Zhou
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Anesthesiology, University of California, San Francisco, 505 Parnassus Ave., Box 0648, San Francisco, CA, USA
| | - Qi Hao
- Department of Anesthesiology, University of California, San Francisco, 505 Parnassus Ave., Box 0648, San Francisco, CA, USA
| | - Shinji Sugita
- Department of Anesthesiology, University of California, San Francisco, 505 Parnassus Ave., Box 0648, San Francisco, CA, USA
| | - Yoshifumi Naito
- Department of Anesthesiology, University of California, San Francisco, 505 Parnassus Ave., Box 0648, San Francisco, CA, USA
| | - Hongli He
- Department of Anesthesiology, University of California, San Francisco, 505 Parnassus Ave., Box 0648, San Francisco, CA, USA
| | - Che-Chung Yeh
- Department of Anesthesiology, University of California, San Francisco, 505 Parnassus Ave., Box 0648, San Francisco, CA, USA
| | - Jae-Woo Lee
- Department of Anesthesiology, University of California, San Francisco, 505 Parnassus Ave., Box 0648, San Francisco, CA, USA.
| |
Collapse
|