1
|
Tsao ST, Gu M, Xiong Q, Deng Y, Deng T, Fu C, Zhao Z, Zhang H, Liu C, Zhong X, Xiang F, Huang F, Wang H. Rapidly Manufactured CAR-T with Conserved Cell Stemness and Distinctive Cytokine-Secreting Profile Shows Improved Anti-Tumor Efficacy. Vaccines (Basel) 2024; 12:1348. [PMID: 39772010 PMCID: PMC11680398 DOI: 10.3390/vaccines12121348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/13/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025] Open
Abstract
Background: The emergence of chimeric antigen receptor T-cell (CAR-T) immunotherapy holds great promise in treating hematologic malignancies. While advancements in CAR design have enhanced therapeutic efficacy, the time-consuming manufacturing process has not been improved in the commercial production of CAR-T cells. In this study, we developed a "DASH CAR-T" process to manufacture CAR-T cells in 72 h and found the excelling anti-tumor efficacy of DASH CAR-T cells over conventionally manufactured CAR-T cells. Methods: Four different CAR-T manufacturing processes were first proposed and examined by flow cytometry in regard to cell viability, T-cell purity and activation, CAR expression, and cell apoptosis. The selected two processes, 48H DASH CAR-T and 72H DASH CAR-T, were applied to the subsequent functional assessments, including T-cell differentiation, antigen-dependent cytotoxicity and expansion, cytokines secretion profile, and in vivo anti-tumor efficacy. Results: We demonstrated that rapidly manufactured CAR-T cells generated within 48-72 h was feasible and exhibited increased naïve and memory T-cell ratios, a distinctive secretory profile, superior expansion capacity, and enhanced in vitro and in vivo anti-tumor activity compared to conventionally manufactured CAR-T cells. Conclusions: Our findings suggest that "DASH CAR-T" process is a valuable platform in reducing CAR-T manufacturing time and producing high-efficacy CAR-T cells for future clinical application.
Collapse
Affiliation(s)
- Shih-Ting Tsao
- Department of R&D, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Mingyuan Gu
- Department of R&D, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Qinghui Xiong
- Department of R&D, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Yingzhi Deng
- Department of R&D, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Tian Deng
- Department of R&D, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Chengbing Fu
- Department of R&D, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Zihao Zhao
- Department of R&D, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Haoyu Zhang
- Department of R&D, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Cuicui Liu
- Department of Regulatory Affairs, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Xiong Zhong
- Department of Medical Research, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Fang Xiang
- Department of Medical Research, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Fei Huang
- Department of R&D, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Haiying Wang
- Department of R&D, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| |
Collapse
|
2
|
Kimura M, Nishikawa K, Imamura J, Kimura K. Stem Cell Growth Factor-β as a Predictive Biomarker of Response to Chemotherapy and Prognosis in Patients with Advanced-Stage Hepatocellular Carcinoma: A Retrospective Study. Cancers (Basel) 2024; 16:320. [PMID: 38254812 PMCID: PMC10813939 DOI: 10.3390/cancers16020320] [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: 11/28/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
In this retrospective study, we investigated the potential application of serum stem cell growth factor beta (SCGF-β) as a biomarker for predicting the therapeutic response and prognosis in patients with hepatocellular carcinoma (HCC) undergoing atezolizumab and bevacizumab (Atz/Bev) combination therapy. Pre- and post-treatment serum SCGF-β levels were measured and analyzed in relation to treatment outcomes and overall survival (OS). Pretreatment SCGF-β levels were associated with treatment response. Patients with SCGF-β levels exceeding the 163,295 pg/mL cutoff experienced significantly reduced OS, with a median OS of 12.03 months, compared to 28.87 months in those with SCGF-β levels at or below this threshold. These findings suggest that SCGF-β can serve as a predictive marker for clinical outcomes in HCC treatment, highlighting the need for prospective studies to further validate these results and clarify the mechanisms underlying SCGF-β-related therapeutic resistance.
Collapse
Affiliation(s)
- Masamichi Kimura
- Department of Hepatology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo 113-8677, Japan; (K.N.); (J.I.); (K.K.)
| | | | | | | |
Collapse
|
3
|
Bolouri H, Ries RE, Wiedeman AE, Hylkema T, Scheiding S, Gersuk VH, O'Brien K, Nguyen QA, Smith JL, Alice Long S, Meshinchi S. Inflammatory bone marrow signaling in pediatric acute myeloid leukemia distinguishes patients with poor outcomes. Nat Commun 2022; 13:7186. [PMID: 36418348 PMCID: PMC9684530 DOI: 10.1038/s41467-022-34965-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 11/09/2022] [Indexed: 11/26/2022] Open
Abstract
High levels of the inflammatory cytokine IL-6 in the bone marrow are associated with poor outcomes in pediatric acute myeloid leukemia (pAML), but its etiology remains unknown. Using RNA-seq data from pre-treatment bone marrows of 1489 children with pAML, we show that > 20% of patients have concurrent IL-6, IL-1, IFNα/β, and TNFα signaling activity and poorer outcomes. Targeted sequencing of pre-treatment bone marrow samples from affected patients (n = 181) revealed 5 highly recurrent patterns of somatic mutation. Using differential expression analyses of the most common genomic subtypes (~60% of total), we identify high expression of multiple potential drivers of inflammation-related treatment resistance. Regardless of genomic subtype, we show that JAK1/2 inhibition reduces receptor-mediated inflammatory signaling by leukemic cells in-vitro. The large number of high-risk pAML genomic subtypes presents an obstacle to the development of mutation-specific therapies. Our findings suggest that therapies targeting inflammatory signaling may be effective across multiple genomic subtypes of pAML.
Collapse
Affiliation(s)
- Hamid Bolouri
- Center for Systems Immunology, Benaroya Research Institute, 1201 9th Ave, Seattle, WA, USA.
| | - Rhonda E Ries
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, USA
| | - Alice E Wiedeman
- Center for Translational Immunology, Benaroya Research Institute, 1201 9th Ave, Seattle, WA, USA
| | - Tiffany Hylkema
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, USA
| | - Sheila Scheiding
- Center for Translational Immunology, Benaroya Research Institute, 1201 9th Ave, Seattle, WA, USA
| | - Vivian H Gersuk
- Center for Systems Immunology, Benaroya Research Institute, 1201 9th Ave, Seattle, WA, USA
| | - Kimberly O'Brien
- Center for Systems Immunology, Benaroya Research Institute, 1201 9th Ave, Seattle, WA, USA
| | - Quynh-Anh Nguyen
- Center for Systems Immunology, Benaroya Research Institute, 1201 9th Ave, Seattle, WA, USA
| | - Jenny L Smith
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, USA
- Research Scientific Computing, Seattle Children's Research Institute, 818 Stewart Street, Seattle, WA, USA
| | - S Alice Long
- Center for Translational Immunology, Benaroya Research Institute, 1201 9th Ave, Seattle, WA, USA
| | - Soheil Meshinchi
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, USA.
| |
Collapse
|
4
|
Daidzein Activates Akt Pathway to Promote the Proliferation of Female Germline Stem Cells through Upregulating Clec11a. Stem Cell Rev Rep 2022; 18:3021-3032. [PMID: 35655001 DOI: 10.1007/s12015-022-10394-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2022] [Indexed: 10/18/2022]
Abstract
Female germline stem cells (FGSCs) have been successfully isolated and characterized from postnatal mammalian and human ovarian tissues. However, the effects and mechanisms of action of natural small-molecule compounds on FGSCs are largely unknown. Here, we found that daidzein promoted the viability and proliferation of FGSCs. To elucidate the mechanism underlying this, we performed RNA-Sequence in daidzein-treated FGSCs and controls. The results showed that there were 153 upregulated and 156 downregulated genes in daidzein treatment. We confirmed the expression of some genes related to cell proliferation in the sequencing results by RT-PCR, such as Type C lectin domain family 11 member a (Clec11a), Mucin1 (Muc1), Glutathione peroxidase 3 (Gpx3), and Tet methylcytosine dioxygenase 1 (Tet1). The high expression of Clec11a at the protein level after daidzein treatment was also confirmed by western blotting. Furthermore, recombinant mouse Clec11a (rmClec11a) protein was shown to promote the viability and proliferation of FGSCs. However, knockdown of Clec11a inhibited the viability and proliferation of FGSCs, which could not be rescued by the administration of daidzein. These results indicate that daidzein promoted the viability and proliferation of FGSCs through Clec11a. In addition, both daidzein and rmClec11a activated the Akt signaling pathway in FGSCs. However, Clec11a knockdown inhibited this pathway, which could not be rescued by daidzein administration. Taken together, our findings revealed that daidzein activates the Akt signaling pathway to promote cell viability and proliferation through upregulating Clec11a. This study should deepen our understanding of the developmental mechanism of FGSCs and female infertility.
Collapse
|
5
|
Lin TY, Yang CH, Chou HC, Cheng CM, Liu YW, Wang JY, Huang LR, Tsai SF, Huang SF, Chen YR. EGFR Mutation-Harboring Lung Cancer Cells Produce CLEC11A with Endothelial Trophic and Tumor-Promoting Activities. Cancers (Basel) 2022; 14:cancers14051356. [PMID: 35267664 PMCID: PMC8909374 DOI: 10.3390/cancers14051356] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Tumor angiogenesis is an important step in the progression of solid tumors. Understanding the mechanisms involved in tumor vasculature formation is critical for improving anti-angiogenic strategies. In this study, we reported that EGFR mutation-containing lung cancer cells produced CLEC11A with endothelial trophic and tumor-promoting activities. CLEC11A could be a novel factor involved in tumor angiogenesis. Abstract The formation of new blood vessels in solid tumors is regulated by various endothelial trophic factors. We identified that CLEC11A, an extracellular C-type lectin, was over-expressed in lung cancer cell lines harboring mutated EGFR. CLEC11A expression was also frequently elevated in lung adenocarcinoma (LAC) tissues with EGFR mutation. CLEC11A-expressing H1299 cells formed larger tumors in nude mice than did the control cells. The CLEC11A-expressing tumors contained more CD31-positive cells, suggesting that they had a higher angiogenic activity. CLEC11A per se did not induce blood vessel formation, but enhanced angiogenesis triggered by VEGF-A or basic FGF in vivo. Additionally, the expression of small hairpin RNA against CLEC11A (shCLEC11A) in HCC827 LAC cells suppressed their tumorigenic ability. Purified CLEC11A exhibited a chemotactic ability, which is dependent on its integrin-binding RGD and LDT motifs, toward endothelial cells. This chemotactic activity was not affected by the presence of a VEGFR inhibitor. Conditioned medium produced by HCC827-shCLEC11A cells had diminished chemotactic ability toward endothelial cells. CLEC11A treatments increased the levels of active integrin β1 that were not associated with activation of focal adhesion kinases in endothelial cells. Our results indicated that CLEC11A was a factor of angiogenic potential and was involved in lung cancer tumorigenesis.
Collapse
Affiliation(s)
- Tzu-Yin Lin
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan 35053, Taiwan; (T.-Y.L.); (C.-H.Y.); (H.-C.C.); (C.-M.C.); (Y.-W.L.); (J.-Y.W.); (L.-R.H.); (S.-F.T.); (S.-F.H.)
| | - Chi-Hwa Yang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan 35053, Taiwan; (T.-Y.L.); (C.-H.Y.); (H.-C.C.); (C.-M.C.); (Y.-W.L.); (J.-Y.W.); (L.-R.H.); (S.-F.T.); (S.-F.H.)
| | - Hsiao-Chin Chou
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan 35053, Taiwan; (T.-Y.L.); (C.-H.Y.); (H.-C.C.); (C.-M.C.); (Y.-W.L.); (J.-Y.W.); (L.-R.H.); (S.-F.T.); (S.-F.H.)
| | - Chun-Mei Cheng
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan 35053, Taiwan; (T.-Y.L.); (C.-H.Y.); (H.-C.C.); (C.-M.C.); (Y.-W.L.); (J.-Y.W.); (L.-R.H.); (S.-F.T.); (S.-F.H.)
| | - Ya-Wen Liu
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan 35053, Taiwan; (T.-Y.L.); (C.-H.Y.); (H.-C.C.); (C.-M.C.); (Y.-W.L.); (J.-Y.W.); (L.-R.H.); (S.-F.T.); (S.-F.H.)
| | - Jiz-Yuh Wang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan 35053, Taiwan; (T.-Y.L.); (C.-H.Y.); (H.-C.C.); (C.-M.C.); (Y.-W.L.); (J.-Y.W.); (L.-R.H.); (S.-F.T.); (S.-F.H.)
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Li-Rung Huang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan 35053, Taiwan; (T.-Y.L.); (C.-H.Y.); (H.-C.C.); (C.-M.C.); (Y.-W.L.); (J.-Y.W.); (L.-R.H.); (S.-F.T.); (S.-F.H.)
| | - Shih-Feng Tsai
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan 35053, Taiwan; (T.-Y.L.); (C.-H.Y.); (H.-C.C.); (C.-M.C.); (Y.-W.L.); (J.-Y.W.); (L.-R.H.); (S.-F.T.); (S.-F.H.)
- Institute of Genetics, Genome Research Center, National Yang-Ming Chiao-Tung University, Hsinchu 30010, Taiwan
| | - Shiu-Feng Huang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan 35053, Taiwan; (T.-Y.L.); (C.-H.Y.); (H.-C.C.); (C.-M.C.); (Y.-W.L.); (J.-Y.W.); (L.-R.H.); (S.-F.T.); (S.-F.H.)
- Department of Anatomic Pathology, Linko Chang Gung Memorial Hospital, Chang-Gung University, Taoyuan 33302, Taiwan
| | - Yi-Rong Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan 35053, Taiwan; (T.-Y.L.); (C.-H.Y.); (H.-C.C.); (C.-M.C.); (Y.-W.L.); (J.-Y.W.); (L.-R.H.); (S.-F.T.); (S.-F.H.)
- Correspondence: ; Tel.:+886-37-206166 (ext. 35311)
| |
Collapse
|
6
|
Tarantino G, Citro V, Balsano C, Capone D. Could SCGF-Beta Levels Be Associated with Inflammation Markers and Insulin Resistance in Male Patients Suffering from Obesity-Related NAFLD? Diagnostics (Basel) 2020; 10:395. [PMID: 32545215 PMCID: PMC7345627 DOI: 10.3390/diagnostics10060395] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 01/08/2023] Open
Abstract
One of the pathologic hallmarks of obesity is macrophage infiltration of adipose tissue that has been confirmed as source of multipotent adult stem cells. Stem cell growth factor-beta (SCGF-β) shows activity on granulocyte/macrophage progenitor cells in combination with granulocyte macrophage colony-stimulating factor (GM-CSF) and macrophage colony-stimulating factor (M-CSF). Obesity-associated inflammation induces insulin resistance (IR), which is central to nonalcoholic fatty liver disease (NAFLD) or hepatic steatosis (HS). We searched for relationship between levels of SCGF-β and those of C-reactive protein (CRP), interleukin-6 (IL-6), tumor necrosis factor-β (TNF-β), interleukin-12p40 (IL-12p40), interleukin-10 (IL-10), ferritin, GM-CSF and M-CSF and between SCGF-β concentrations and IR in obese patients with HS. Eighty obese patients were retrospectively studied. Serum cytokines levels were appreciated by magnetic bead-based multiplex immunoassays. IR was evaluated by homeostatic model assessment (HOMA), HOMA-derived β-cell function (HOMA-B%), quantitative insulin sensitivity check Index (QUICKI) and single point insulin sensitivity estimator (SPISE). HS and spleen volume were assessed by ultrasonography (US). SCGF-β and IL-6 levels predicted HOMA values (p = 0.032 and 0.041, respectively) only in males. In male patients, CRP and IL-6 levels (p = 0.007) predicted SCGF-β concentrations (p = 0.03 and 0.007, respectively), which in turn predicted HS at US, p = 0.037. SCGF-β levels were linked to IR and HS severity with the mediation role of CRP. IL-10 levels negatively predicted SCGF-β concentrations (p = 0.033). M-CSF levels predicted serum concentration of both TNF-β and IL-12p40 (p = 0.00), but did not predict serum IL-10 (p = 0.30). Prediction of HOMA values by SCGF-β levels, likely mediated by markers of inflammation, characterizes this study, shedding some light on mechanisms inducing/worsening IR of male patients with obesity-related NAFLD.
Collapse
Affiliation(s)
- Giovanni Tarantino
- Department of Clinical Medicine and Surgery, Federico II University Medical School of Naples, 80131 Naples, Italy
| | - Vincenzo Citro
- Department of General Medicine, “Umberto I” Hospital, Nocera Inferiore (Sa), 84014 Nocera Inferiore, Italy;
| | - Clara Balsano
- Department of Clinical Medicine, Life, Health & Environmental Sciences-MESVA, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Domenico Capone
- Clinical Pharmacology Consultant, Casoria, 80026 Naples, Italy;
| |
Collapse
|
7
|
Wang M, Guo J, Zhang L, Kuek V, Xu J, Zou J. Molecular structure, expression, and functional role of Clec11a in skeletal biology and cancers. J Cell Physiol 2020; 235:6357-6365. [PMID: 32003015 DOI: 10.1002/jcp.29600] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 01/13/2020] [Indexed: 12/20/2022]
Abstract
C-type lectin domain family 11 member A (Clec11a), also known as stem cell growth factor (SCGF), C-type lectin superfamily member 3 (CLECSF3), or osteolectin was initially identified as a growth factor for hematopoietic progenitor cells. The human Clec11a gene encodes a polypeptide of 323 amino acids with characteristics of a secreted glycoprotein encompassing two integrin-binding motifs, RGD (Arg-Gly-Asp) and LDT (Leu-Asp-Thr), a putative leucine zipper domain, and a functional C-type lectin domain. It regulates hematopoietic differentiation and homeostasis and exhibits a protective effect against severe malarial anemia and lipotoxicity. Furthermore, Clec11a promotes the differentiation of mesenchymal progenitors into mature osteoblasts in vitro and plays an important role in the maintenance of adult skeleton age-related bone loss and fracture repair. Receptor ligand binding results in activation of downstream signaling cascades including glycogen synthase kinase 3 (GSK3), β-catenin, and Wnt, resulting in the expression of osteoblast-related gene transcripts including Alp, Runx2, Lef1, and Axin2. In addition, Clec11a is also associated with the development of several cancers, including leukemia, multiple myeloma, and gastrointestinal tract tumors. To date, however, the mechanisms governing transcription regulation of the Clec11a gene are not known and remain to be uncovered. Understanding the function and mechanism of action of Clec11a will pave the way for the development of Clec11a as a novel therapeutic target for conditions such as cancer, anemia, and skeletal diseases.
Collapse
Affiliation(s)
- Miao Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Jianmin Guo
- School of Kinesiology, Shanghai University of Sport, Shanghai, China.,School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Lingli Zhang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China.,School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Vincent Kuek
- School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Jiake Xu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China.,School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Jun Zou
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
8
|
Efremov YR, Proskurina AS, Potter EA, Dolgova EV, Efremova OV, Taranov OS, Ostanin AA, Chernykh ER, Kolchanov NA, Bogachev SS. Cancer Stem Cells: Emergent Nature of Tumor Emergency. Front Genet 2018; 9:544. [PMID: 30505319 PMCID: PMC6250818 DOI: 10.3389/fgene.2018.00544] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/26/2018] [Indexed: 12/12/2022] Open
Abstract
A functional analysis of 167 genes overexpressed in Krebs-2 tumor initiating cells was performed. In the first part of the study, the genes were analyzed for their belonging to one or more of the three groups, which represent the three major phenotypic manifestation of malignancy of cancer cells, namely (1) proliferative self-sufficiency, (2) invasive growth and metastasis, and (3) multiple drug resistance. 96 genes out of 167 were identified as possible contributors to at least one of these fundamental properties. It was also found that substantial part of these genes are also known as genes responsible for formation and/or maintenance of the stemness of normal pluri-/multipotent stem cells. These results suggest that the malignancy is simply the ability to maintain the stem cell specific genes expression profile, and, as a consequence, the stemness itself regardless of the controlling effect of stem niches. In the second part of the study, three stress factors combined into the single concept of "generalized cellular stress," which are assumed to activate the expression of these genes, were defined. In addition, possible mechanisms for such activation were identified. The data obtained suggest the existence of a mechanism for the de novo formation of a pluripotent/stem phenotype in the subpopulation of "committed" tumor cells.
Collapse
Affiliation(s)
- Yaroslav R Efremov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia.,Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Anastasia S Proskurina
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Ekaterina A Potter
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Evgenia V Dolgova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Oksana V Efremova
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Oleg S Taranov
- The State Research Center of Virology and Biotechnology Vector, Koltsovo, Russia
| | - Aleksandr A Ostanin
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Elena R Chernykh
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Nikolay A Kolchanov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Sergey S Bogachev
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
9
|
Sukowati CHC, Patti R, Pascut D, Ladju RB, Tarchi P, Zanotta N, Comar M, Tiribelli C, Crocè LS. Serum Stem Cell Growth Factor Beta for the Prediction of Therapy Response in Hepatocellular Carcinoma. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6435482. [PMID: 30246025 PMCID: PMC6136561 DOI: 10.1155/2018/6435482] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 07/11/2018] [Accepted: 08/02/2018] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Chronic inflammatory response is one of major contributors in the development of hepatocellular carcinoma (HCC). Inflammatory molecules, such as cytokines and growth factors in the circulation, can be useful in the diagnosis and prognosis of the patients. The stem cell growth factor beta (SCGFβ), a newly found protein, is a secreted sulfated glycoprotein and it functions as a growth factor for primitive hematopoietic progenitor cells. The level of SCGFβ had been reported to be elevated in several cancer types. However, there is very few or even no information on this protein in the study of HCC, even more in clinical studies. METHODS A multiplex immunoassay panel of 48 cytokines and growth factors were utilized to screen 68 sera from 29 HCC patients at pretreatment (T0), 1 month (T1), and 6 months (T6) after treatment by either radiofrequency ablation (RF) or transarterial chemoembolization (TACE). Treatment response was evaluated according to mRECIST criteria. RESULTS Immunoassay screening showed that the levels of IL-17, CTACK, TNFα, IL-2Rα, IL-8, and SCGFβ were different in Complete Responders (CR) and Nonresponders (NR) groups. At T0 and T1, the SCGFβ level was significantly the highest in NR (23.8 and 40.7 ng/mL, respectively), followed by early recurrence (25.4 and 25.0 ng/mL), and CR (6.7 and 5.3 ng/mL), independently from HCV, stages, and treatment type. Low basal SCGFβ level was associated with longer disease-free survival compared to high SCGFβ. CONCLUSION In this study, for the first time, we demonstrate that the high level of serum SCGFβ at pre- and posttreatment is associated with HCC nonresponsiveness.
Collapse
Affiliation(s)
| | - Riccardo Patti
- Fondazione Italiana Fegato, AREA Science Park Basovizza, Trieste, Italy
- Department of Medicine, Surgery, and Health Sciences, University of Trieste, Trieste, Italy
| | - Devis Pascut
- Fondazione Italiana Fegato, AREA Science Park Basovizza, Trieste, Italy
| | - Rusdina B. Ladju
- Fondazione Italiana Fegato, AREA Science Park Basovizza, Trieste, Italy
- Faculty of Medicine, University of Hasanuddin, Makassar, Indonesia
| | | | - Nunzia Zanotta
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Manola Comar
- Department of Medicine, Surgery, and Health Sciences, University of Trieste, Trieste, Italy
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Claudio Tiribelli
- Fondazione Italiana Fegato, AREA Science Park Basovizza, Trieste, Italy
| | - Lory S. Crocè
- Fondazione Italiana Fegato, AREA Science Park Basovizza, Trieste, Italy
- Department of Medicine, Surgery, and Health Sciences, University of Trieste, Trieste, Italy
- Teaching Hospital, ASUITS, Trieste, Italy
| |
Collapse
|
10
|
Li Y, Zhao H, Xu Q, Lv N, Jing Y, Wang L, Wang X, Guo J, Zhou L, Liu J, Chen G, Chen C, Li Y, Yu L. Detection of prognostic methylation markers by methylC-capture sequencing in acute myeloid leukemia. Oncotarget 2017; 8:110444-110459. [PMID: 29299160 PMCID: PMC5746395 DOI: 10.18632/oncotarget.22789] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 11/15/2017] [Indexed: 12/11/2022] Open
Abstract
Clinical and genetic features incompletely predict outcome in acute myeloid leukemia (AML). The value of clinical methylation assays for prognostic markers has not been extensively explored. We assess the prognostic implications of methylC-capture sequencing (MCC-Seq) in patients with de novo AML by integrating DNA methylation and genetic risk stratification. MCC-Seq assessed DNA methylation level in 44 samples. The differentially methylated regions associated with prognostic genetic information were identified. The selected prognostic DNA methylation markers were independently validated in two sets. MCC-Seq exhibited good performance in AML patients. A panel of 12 differentially methylated genes was identified with promoter hyper-differentially methylated regions associated with the outcome. Compared with a low M-value, a high M-value was associated with failure to achieve complete remission (p = 0.024), increased hazard for disease-free survival in the study set (p = 0.039) and poor overall survival in The Cancer Genome Atlas set (p = 0.038). Hematopoietic stem cell transplantation and survival outcomes were not adversely affected by a high M-value (p = 0.271). Our study establishes that MCC-Seq is a stable, reproducible, and cost-effective methylation assay in AML. A 12-gene M-value encompassing epigenetic and genetic prognostic information represented a valid prognostic marker for patients with AML.
Collapse
Affiliation(s)
- Yan Li
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing 100853, China.,Department of Hematology, Hainan Branch of Chinese PLA General Hospital, Sanya 572013, China
| | - Hongmei Zhao
- Annoroad Gene Technology Co. Ltd., Beijing 100176, China
| | - Qingyu Xu
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing 100853, China.,Medical School of Nankai University, Tianjin 300071, China
| | - Na Lv
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing 100853, China.,Department of Hematology, General Hospital of Shenzhen University, Shenzhen 518060, China
| | - Yu Jing
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Lili Wang
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiaowen Wang
- Annoroad Gene Technology Co. Ltd., Beijing 100176, China
| | - Jing Guo
- Annoroad Gene Technology Co. Ltd., Beijing 100176, China
| | - Lei Zhou
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Jing Liu
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Guofeng Chen
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing 100853, China.,Medical School of Nankai University, Tianjin 300071, China
| | - Chongjian Chen
- Annoroad Gene Technology Co. Ltd., Beijing 100176, China
| | - Yonghui Li
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Li Yu
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing 100853, China.,Department of Hematology, General Hospital of Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
11
|
Jiang Y, Xu P, Yao D, Chen X, Dai H. CD33, CD96 and Death Associated Protein Kinase (DAPK) Expression Are Associated with the Survival Rate and/or Response to the Chemotherapy in the Patients with Acute Myeloid Leukemia (AML). Med Sci Monit 2017; 23:1725-1732. [PMID: 28391288 PMCID: PMC5395139 DOI: 10.12659/msm.900305] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Background Leukemia stem cells (LSC) are involved in the incidence, drug resistance, and relapse of leukemia while LSC-related antigen CD33, CD96, and DAPK expression in AML and its prognosis is still unclear. This study explored LSC-related antigens expression in acute myeloid leukemia (AML) and its prognosis. Material/Methods A total of 156 cases of AML patients were enrolled in the experiment. The expression of CD33, CD96, and DAPK in CD34+CD38−CD123+ LSC were tested by flow cytometry. The survival curve was established using the Kaplan-Meier method. Results Among different subtypes of AML, the positive rate of CD33 was M3> M5> M1> M2> M4; for CD96 it was M5> M4> M2> M3> M1; and for DAPK it was M3> M2> M5> M4> M1. After chemotherapy, the response rate in CD33 and CD96 high expression groups, and DAPK low expression group was significantly lower than the groups with CD33 low expression, CD96 low expression, and DAPK high expression. The median survival time in the CD33 high expression group was markedly lower than the CD33 low expression group (36.5 months). The CD96 high expression group exhibited obviously shorter median survival time than the CD96 low expression group. The DAPK high expression group exhibited longer median survival time than the DAPK low expression group. Conclusions CD33 and CD96 overexpression, and DAPK downregulation in the LSC of AML patients were associated with poor chemotherapy effect and prognosis, and higher recurrence rate.
Collapse
Affiliation(s)
- Yongfang Jiang
- Department of Hematology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Ping Xu
- Department of Hematology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Dandan Yao
- Department of Hematology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Xi Chen
- Department of Hematology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Haibin Dai
- Department of Hematology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| |
Collapse
|
12
|
Da Riva L, Bozzi F, Mondellini P, Miccichè F, Fumagalli E, Vaghi E, Tarantino E, Huber V, Gronchi A, Tamborini E, Pierotti MA, Pilotti S, Bongarzone I. Proteomic detection of a large amount of SCGFα in the stroma of GISTs after imatinib therapy. J Transl Med 2011; 9:158. [PMID: 21943129 PMCID: PMC3192683 DOI: 10.1186/1479-5876-9-158] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 09/23/2011] [Indexed: 01/24/2023] Open
Abstract
Background Gastrointestinal stromal tumors (GISTs) are the most frequent mesenchymal tumors to develop in the digestive tract. These tumors are highly resistant to conventional chemotherapy and only the introduction of imatinib mesylate has improved the prognosis of patients. However, Response Evaluation Criteria in Solid Tumors are inappropriate for assessing tumor response, and the histological/pathological response to imatinib is variable, heterogeneous, and does not associate with clinical response. The effects of imatinib on responding GISTs are still being explored, and few studies correlate the clinical response with the histological response after pharmacological treatment. Recently, apoptosis and autophagy were suggested as possible alternative mechanisms of pharmacological response. Methods Here, we used a proteomic approach, combined with other analyses, to identify some molecular stromal components related to the response/behavior of resected, high-risk GISTs after neoadiuvant imatinib therapy. Results Our proteomic results indicate an elevated concentration of Stem Cell Growth Factor (SCGF), a hematopoietic growth factor having a role in the development of erythroid and myeloid progenitors, in imatinib-responsive tumor areas. SCGFα expression was detected by mass spectrometry, immunohistochemistry and/or western blot and attributed to acellular matrix of areas scored negative for KIT (CD117). RT-PCR results indicated that GIST samples did not express SCGF transcripts. The recently reported demonstration by Gundacker et al. [1] of the secretion of SCGF in mature pro-inflammatory dendritic cells would indicate a potential importance of SCGF in tissue inflammatory response. Accordingly, inflammatory infiltrates were detected in imatinib-affected areas and the CD68-positivity of the SCGF-positive and KIT-negative areas suggested previous infiltration of monocytes/macrophages into these regions. Thus, chronic inflammation subsequent to imatinib treatment may determine monocyte/macrophage recruitment in imatinib-damaged areas; these areas also feature prominent tumor-cell loss that is replaced by dense hyalinization and fibrosis. Conclusions Our studies highlight a possible role of SCGFα in imatinib-induced changes of GIST structure, consistent with a therapeutic response.
Collapse
Affiliation(s)
- Luca Da Riva
- Proteomics Laboratory, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Penolazzi L, Tavanti E, Vecchiatini R, Lambertini E, Vesce F, Gambari R, Mazzitelli S, Mancuso F, Luca G, Nastruzzi C, Piva R. Encapsulation of mesenchymal stem cells from Wharton's jelly in alginate microbeads. Tissue Eng Part C Methods 2010; 16:141-55. [PMID: 19402785 DOI: 10.1089/ten.tec.2008.0582] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The description of a microencapsulation procedure for Wharton's jelly mesenchymal stem cells (WJMSCs) is reported. The applied method is based on the generation of monodisperse droplets by a vibrational nozzle. An ionic alginate encapsulation procedure was utilized for the microbeads hardening. Different experimental parameters were analyzed, including frequency and amplitude of vibration, polymer pumping rate, and distance between the nozzle and the gelling bath. The produced barium-alginate microbeads were characterized by excellent morphological characteristics as well as a very narrow size distribution. The microencapsulation procedure did not alter the morphology and viability of the encapsulated WJMSCs. In addition, the current paper reports the functional properties in terms of secretive profiles of both free and encapsulated WJMSCs. The analyzed factors were members of the family of interleukins, chemokines, growth factors, and soluble forms of adhesion molecules. These experiments showed that despite encapsulation, most of the proteins analyzed were secreted both by the free and encapsulated cells, even if in a different extent. In conclusion, the described encapsulation procedure represents a promising strategy to utilize WJMSCs for possible in vivo applications in tissue engineering and biomedicine.
Collapse
Affiliation(s)
- Letizia Penolazzi
- Department of Biochemistry and Molecular Biology, University of Ferrara , Ferrara, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|