1
|
Hartman SJ, Hibberd MC, Mostafa I, Naila NN, Islam MM, Zaman MU, Huq S, Mahfuz M, Islam MT, Mukherji K, Moghaddam VA, Chen RY, Province MA, Webber DM, Henrissat S, Henrissat B, Terrapon N, Rodionov DA, Osterman AL, Barratt MJ, Ahmed T, Gordon JI. A microbiome-directed therapeutic food for children recovering from severe acute malnutrition. Sci Transl Med 2024; 16:eadn2366. [PMID: 39356745 DOI: 10.1126/scitranslmed.adn2366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 07/22/2024] [Indexed: 10/04/2024]
Abstract
Globally, severe acute malnutrition (SAM), defined as a weight-for-length z-score more than three SDs below a reference mean (WLZ < -3), affects 14 million children under 5 years of age. Complete anthropometric recovery after standard, short-term interventions is rare, with children often left with moderate acute malnutrition (MAM; WLZ -2 to -3). We conducted a randomized controlled trial (RCT) involving 12- to 18-month-old Bangladeshi children from urban and rural sites, who, after initial hospital-based treatment for SAM, received a 3-month intervention with a microbiome-directed complementary food (MDCF-2) or a calorically more dense, standard ready-to-use supplementary food (RUSF). The rate of WLZ improvement was significantly greater in MDCF-2-treated children (P = 8.73 × 10-3), similar to our previous RCT of Bangladeshi children with MAM without antecedent SAM (P = 0.032). A correlated meta-analysis of plasma levels of 4520 proteins in both RCTs revealed 215 positively associated with WLZ (largely representing musculoskeletal and central nervous system development) and 44 negatively associated (primarily related to immune activation). Moreover, the positively associated proteins were significantly enriched by MDCF-2 (q = 1.1 × 10-6). Characterizing the abundances of 754 bacterial metagenome-assembled genomes in serially collected fecal samples disclosed the effects of acute rehabilitation for SAM on the microbiome and how, during treatment for MAM, specific strains of Prevotella copri function at the intersection between MDCF-2 glycan metabolism and anthropometric recovery. These results provide a rationale for further testing the generalizability of MDCF efficacy and for identifying biomarkers to define treatment responses.
Collapse
Affiliation(s)
- Steven J Hartman
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Newman Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Matthew C Hibberd
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Newman Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ishita Mostafa
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Nurun N Naila
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Md Munirul Islam
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Mahabub Uz Zaman
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Sayeeda Huq
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Mustafa Mahfuz
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Md Tazul Islam
- Terre des Hommes Netherlands - Bangladesh Country Office, Dhaka 1209, Bangladesh
| | - Kallol Mukherji
- Terre des Hommes Netherlands - Bangladesh Country Office, Dhaka 1209, Bangladesh
| | - Vaha Akbary Moghaddam
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Robert Y Chen
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Newman Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Daniel M Webber
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Newman Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Suzanne Henrissat
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Newman Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bernard Henrissat
- Department of Biotechnology and Biomedicine (DTU Bioengineering), Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| | - Nicolas Terrapon
- Architecture et Fonction des Macromolécules Biologiques, CNRS, Aix-Marseille University, F-13288 Marseille, France
| | - Dmitry A Rodionov
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Andrei L Osterman
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Michael J Barratt
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Newman Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tahmeed Ahmed
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Jeffrey I Gordon
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Newman Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
2
|
Zhang L, Pan Y, Pan F, Huang S, Wang F, Zeng Z, Chen H, Tian X. MATN4 as a target gene of HIF-1α promotes the proliferation and metastasis of osteosarcoma. Aging (Albany NY) 2024; 16:10462-10476. [PMID: 38889378 PMCID: PMC11236324 DOI: 10.18632/aging.205941] [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: 08/28/2023] [Accepted: 03/03/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Osteosarcoma is a highly malignant bone tumor that exhibits rapid growth and early metastasis. Hypoxia plays a pivotal role in promoting the proliferation and metastasis of osteosarcoma through a series of molecular events, which are partially mediated and regulated by HIF-1α. However, the regulatory network associated with HIF-1α in osteosarcoma remains limited. Therefore, the objective of this study was to identify critical hypoxia-associated genes and investigate their effects and molecular mechanisms in osteosarcoma cells. METHODS Through bioinformatics analysis, matrilin-4 (MATN4) was identified as a crucial gene associated with hypoxia. The expression of MATN4 and HIF-1α was assessed using immunohistochemistry, RT-qPCR, and western blotting. The proliferative capacity of osteosarcoma cells was assessed through the utilization of CCK-8, EDU staining, and colony formation assays. The effects of MATN4 on the mobility of OS cells were evaluated using wound-healing assays and transwell assays. The interaction between MATN4 and HIF-1α was detected through chromatin immunoprecipitation. RESULTS MATN4 is overexpressed in osteosarcoma tissue and cells, particularly in osteosarcoma cells with high metastatic potential. Knockdown of MATN4 inhibits the proliferation, migration, and invasion abilities of osteosarcoma cells and reverses the promoting effects of hypoxia on these functions. Additionally, HIF-1α binds to MATN4 and upregulates its expression. Interestingly, knockdown of HIF-1α reduces the stimulatory effects of MATN4 overexpression on the proliferation, migration, and invasion of osteosarcoma cells under hypoxic conditions. CONCLUSIONS Taken together, our results suggest that MATN4 is regulated by HIF-1α and confers a more aggressive phenotype on OS cells. This evidence suggests that MATN4 may act as a potential target for OS diagnosis and treatment.
Collapse
Affiliation(s)
- Lu Zhang
- School of Clinical Medicine, Guizhou Medical University, Guiyang 550000, Guizhou, China
| | - Yujie Pan
- Department of Orthopedics, The Affiliated Hospital of Guizhou Medical University, Guiyang 550000, Guizhou, China
- Department of Traumatic Orthopedics, The Affiliated Hospital of Guizhou Medical University, Guiyang 550000, Guizhou, China
| | - Feng Pan
- Department of Bone and Joint Surgery, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang 550000, Guizhou, China
| | - Songsong Huang
- Department of Pathology, The Afflicted Hospital of Guizhou Medical University, Guiyang 550000, Guizhou, China
| | - Fengyan Wang
- School of Clinical Medicine, Guizhou Medical University, Guiyang 550000, Guizhou, China
| | - Zhirui Zeng
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Guizhou Medical University, Guiyang 550000, China
| | - Houping Chen
- Department of Orthopedics, Guiyang Maternal and Child Health-Care Hospital, Guiyang 550000, China
| | - Xiaobin Tian
- School of Clinical Medicine, Guizhou Medical University, Guiyang 550000, Guizhou, China
- Department of Orthopedics, The Affiliated Hospital of Guizhou Medical University, Guiyang 550000, Guizhou, China
| |
Collapse
|
3
|
Zhou X, Zhu Y, Gao D, Li M, Lin L, Wang Z, Du H, Xu Y, Liu J, He Y, Guo Y, Wang S, Qiao S, Bao Y, Liu Y, Zhang H. Matrilin-3 supports neuroprotection in ischemic stroke by suppressing astrocyte-mediated neuroinflammation. Cell Rep 2024; 43:113980. [PMID: 38520693 DOI: 10.1016/j.celrep.2024.113980] [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: 05/05/2020] [Revised: 02/08/2024] [Accepted: 03/06/2024] [Indexed: 03/25/2024] Open
Abstract
In the brain, the role of matrilin-3, an extracellular matrix component in cartilage, is unknown. Here, we identify that matrilin-3 decreased in reactive astrocytes but was unchanged in neurons after ischemic stroke in animals. Importantly, it declined in serum of patients with acute ischemic stroke. Genetic or pharmacological inhibition or supplementation of matrilin-3 aggravates or reduces brain injury, astrocytic cell death, and glial scar, respectively, but has no direct effect on neuronal cell death. RNA sequencing demonstrates that Matn3-/- mice display an increased inflammatory response profile in the ischemic brain, including the nuclear factor κB (NF-κB) signaling pathway. Both endogenous and exogenous matrilin-3 reduce inflammatory mediators. Mechanistically, extracellular matrilin-3 enters astrocytes via caveolin-1-mediated endocytosis. Cytoplasmic matrilin-3 translocates into the nucleus by binding to NF-κB p65, suppressing inflammatory cytokine transcription. Extracellular matrilin-3 binds to BMP-2, blocking the BMP-2/Smads pathway. Thus, matrilin-3 is required for astrocytes to exert neuroprotection, at least partially, by suppressing astrocyte-mediated neuroinflammation.
Collapse
Affiliation(s)
- Xianyong Zhou
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, Suzhou Key Laboratory of Drug Research for Prevention and Treatment of Hyperlipidemic Diseases, Suzhou International Joint Laboratory for Diagnosis and Treatment of Brain Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yongming Zhu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, Suzhou Key Laboratory of Drug Research for Prevention and Treatment of Hyperlipidemic Diseases, Suzhou International Joint Laboratory for Diagnosis and Treatment of Brain Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215123, China
| | - Defei Gao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, Suzhou Key Laboratory of Drug Research for Prevention and Treatment of Hyperlipidemic Diseases, Suzhou International Joint Laboratory for Diagnosis and Treatment of Brain Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215123, China
| | - Min Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, Suzhou Key Laboratory of Drug Research for Prevention and Treatment of Hyperlipidemic Diseases, Suzhou International Joint Laboratory for Diagnosis and Treatment of Brain Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215123, China
| | - Liang Lin
- The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, China
| | - Zhanxiang Wang
- The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, China
| | - Huaping Du
- Department of Neurology, Suzhou Ninth People's Hospital, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, Jiangsu 215200, China
| | - Yuan Xu
- Department of Neurology, Suzhou Ninth People's Hospital, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, Jiangsu 215200, China
| | - Jin Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, Suzhou Key Laboratory of Drug Research for Prevention and Treatment of Hyperlipidemic Diseases, Suzhou International Joint Laboratory for Diagnosis and Treatment of Brain Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yang He
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, Suzhou Key Laboratory of Drug Research for Prevention and Treatment of Hyperlipidemic Diseases, Suzhou International Joint Laboratory for Diagnosis and Treatment of Brain Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yi Guo
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, Suzhou Key Laboratory of Drug Research for Prevention and Treatment of Hyperlipidemic Diseases, Suzhou International Joint Laboratory for Diagnosis and Treatment of Brain Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215123, China
| | - Shuai Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, Suzhou Key Laboratory of Drug Research for Prevention and Treatment of Hyperlipidemic Diseases, Suzhou International Joint Laboratory for Diagnosis and Treatment of Brain Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215123, China
| | - Shigang Qiao
- Kunshan Hospital of Chinese Medicine, Affiliated Hospital of Yangzhou University, Suzhou, Jiangsu 215301, China; Suzhou Science & Technology Town Hospital, Suzhou, Jiangsu 215163, China
| | - Yingshi Bao
- Department of Neurology, Suzhou Ninth People's Hospital, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, Jiangsu 215200, China
| | - Yuan Liu
- Department of Neurology, Suzhou Ninth People's Hospital, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, Jiangsu 215200, China.
| | - Huiling Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, Suzhou Key Laboratory of Drug Research for Prevention and Treatment of Hyperlipidemic Diseases, Suzhou International Joint Laboratory for Diagnosis and Treatment of Brain Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
4
|
Chen H, Guo S, Li R, Yang L, Wang R, Jiang Y, Hao Y. YTHDF2-regulated matrilin-3 mitigates post-reperfusion hemorrhagic transformation in ischemic stroke via the PI3K/AKT pathway. J Neuropathol Exp Neurol 2024; 83:194-204. [PMID: 38230623 PMCID: PMC10880072 DOI: 10.1093/jnen/nlad102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024] Open
Abstract
Hemorrhagic transformation can complicate ischemic strokes after recanalization treatment within a time window that requires early intervention. To determine potential therapeutic effects of matrilin-3, rat cerebral ischemia-reperfusion was produced using transient middle cerebral artery occlusion (tMCAO); intracranial hemorrhage and infarct volumes were assayed through hemoglobin determination and 2,3,5-triphenyltetrazoliumchloride (TTC) staining, respectively. Oxygen-glucose deprivation (OGD) modeling of ischemia was performed on C8-D1A cells. Interactions between matrilin-3 and YTH N6-methyladenosine RNA binding protein F2 (YTHDF2) were determined using RNA immunoprecipitation assay and actinomycin D treatment. Reperfusion after tMCAO modeling increased hemorrhage, hemoglobin content, and infarct volumes; these were alleviated by matrilin treatment. Matrilin-3 was expressed at low levels and YTHDF2 was expressed at high levels in ischemic brains. In OGD-induced cells, matrilin-3 was negatively regulated by YTHDF2. Matrilin-3 overexpression downregulated p-PI3K/PI3K, p-AKT/AKT, ZO-1, VE-cadherin and occludin, and upregulated p-JNK/JNK in ischemic rat brains; these effects were reversed by LY294002 (a PI3K inhibitor). YTHDF2 knockdown inactivated the PI3K/AKT pathway, inhibited inflammation and decreased blood-brain barrier-related protein levels in cells; these effects were reversed by matrilin-3 deficiency. These results indicate that YTHDF2-regulated matrilin-3 protected ischemic rats against post-reperfusion hemorrhagic transformation via the PI3K/AKT pathway and that matrilin may have therapeutic potential in ischemic stroke.
Collapse
Affiliation(s)
- Hanze Chen
- Department of Neurology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, China
| | - Siping Guo
- Department of Neurology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou City, Jiangsu Province, China
| | - Runnan Li
- Department of Neurology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou City, Jiangsu Province, China
| | - Lihui Yang
- Department of Neurology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou City, Jiangsu Province, China
| | - Rui Wang
- Department of Neurology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou City, Jiangsu Province, China
| | - Yasi Jiang
- Department of Neurology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou City, Jiangsu Province, China
| | - Yonggang Hao
- Department of Neurology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, China
- Department of Neurology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou City, Jiangsu Province, China
| |
Collapse
|
5
|
Huang Y, Xu X, Lu Y, Sun Q, Zhang L, Shao J, Chen D, Chang Y, Sun X, Zhuo W, Zhou T. The phase separation of extracellular matrix protein matrilin-3 from cancer-associated fibroblasts contributes to gastric cancer invasion. FASEB J 2024; 38:e23406. [PMID: 38193601 DOI: 10.1096/fj.202301524r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/23/2023] [Accepted: 12/19/2023] [Indexed: 01/10/2024]
Abstract
Cancer-associated fibroblast (CAF) has emerged as a key contributor to the remodeling of tumor microenvironment through the expression and secretion of extracellular matrix (ECM) proteins, thereby promoting carcinogenesis. However, the precise contribution of ECM proteins from CAFs to gastric carcinogenesis remains poorly understood. In this study, we find that matrilin-3 (MATN3), an upregulated ECM protein associated with poorer prognosis in gastric cancer patients, originates from CAFs in gastric cancer tissues. Ectopic expression of MATN3 in CAFs significantly promotes the invasion of gastric cancer cells, which can be attenuated by neutralizing MATN3 with its antibody. Notably, a portion of MATN3 protein is found to form puncta in gastric cancer tissues ECM. MATN3 undergoes phase separation, which is mediated by its low complexity (LC) and coiled-coil (CC) domains. Moreover, overexpression of MATN3 deleted with either LC or CC in CAFs is unable to promote the invasion of gastric cancer cells, suggesting that LC or CC domain is required for the effect of CAF-secreted MATN3 in gastric cancer cell invasion. Additionally, orthotopic co-injection of gastric cancer cells and CAFs expressing MATN3, but not its ΔLC and ΔCC mutants, leads to enhanced gastric cancer cell invasion in mouse models. Collectively, our works suggest that MATN3 is secreted by CAFs and undergoes phase separation, which promotes gastric cancer invasion.
Collapse
Affiliation(s)
- Yuliang Huang
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyang Xu
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Yunkun Lu
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiang Sun
- Center for RNA Medicine, International Institutes of Medicine and the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Lu Zhang
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaqi Shao
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Dingwei Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongxia Chang
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoxia Sun
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Zhuo
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Tianhua Zhou
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, China
- Zhejiang University Cancer Center, Hangzhou, China
| |
Collapse
|
6
|
Toyota K, Akashi H, Ishikawa M, Yamaguchi K, Shigenobu S, Sato T, Lange A, Tyler CR, Iguchi T, Miyagawa S. Comparative analysis of gonadal transcriptomes between turtle and alligator identifies common molecular cues activated during the temperature-sensitive period for sex determination. Gene 2023; 888:147763. [PMID: 37666375 DOI: 10.1016/j.gene.2023.147763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/09/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023]
Abstract
The mode of sex determination in vertebrates can be categorized as genotypic or environmental. In the case of genotypic sex determination (GSD), the sexual fate of an organism is determined by the chromosome composition with some having dominant genes, named sex-determining genes, that drive the sex phenotypes. By contrast, many reptiles exhibit environmental sex determination (ESD), whereby environmental stimuli drive sex determination, and most notably temperature. To date, temperature-dependent sex determination (TSD) has been found in most turtles, some lizards, and all crocodylians, but commonalities in the controlling processes are not well established. Recent innovative sequencing technology has enabled investigations into gonadal transcriptomic profiles during temperature-sensitive periods (TSP) in various TSD species which can help elucidate the controlling mechanisms. In this study, we conducted a time-course analysis of the gonadal transcriptome during the male-producing temperature (26℃) of the Reeve's turtle (Chinese three-keeled pond turtle) Mauremys reevesii. We then compared the transcriptome profiles for this turtle species during the TSP with that for the American alligator Alligator mississippiensis to identify conserved reptilian TSD-related genes. Our transcriptome-based findings provide an opportunity to retrieve the candidate molecular cues that are activated during TSP and compare these target responses between TSD and GSD turtle species, and between TSD species.
Collapse
Affiliation(s)
- Kenji Toyota
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo 125-8585, Japan; Noto Marine Laboratory, Institute of Nature and Environmental Technology, Kanazawa University, Ogi, Noto-cho, Ishikawa 927-0553, Japan; Department of Biological Sciences, Faculty of Science, Kanagawa University, 2946 Tsuchiya, Hiratsuka, Kanagawa 259-1293, Japan.
| | - Hiroshi Akashi
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo 125-8585, Japan; Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-0882, Japan
| | - Momoka Ishikawa
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo 125-8585, Japan
| | - Katsushi Yamaguchi
- Trans-Omics Facility, National Institute for Basic Biology, 38 Nishigonaka, Myodaiji-cho, Okazaki, Aichi 444-8585, Japan
| | - Shuji Shigenobu
- Trans-Omics Facility, National Institute for Basic Biology, 38 Nishigonaka, Myodaiji-cho, Okazaki, Aichi 444-8585, Japan
| | - Tomomi Sato
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan
| | - Anke Lange
- Biosciences, Faculty of Health and Life Sciences, University of Exeter, Stocker Road, Exeter EX4 4QD, UK
| | - Charles R Tyler
- Biosciences, Faculty of Health and Life Sciences, University of Exeter, Stocker Road, Exeter EX4 4QD, UK
| | - Taisen Iguchi
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Kanazawa University, Ogi, Noto-cho, Ishikawa 927-0553, Japan; Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan
| | - Shinichi Miyagawa
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo 125-8585, Japan.
| |
Collapse
|
7
|
Matera I, Miglionico R, Abruzzese V, Marchese G, Ventola GM, Castiglione Morelli MA, Bisaccia F, Ostuni A. A Regulator Role for the ATP-Binding Cassette Subfamily C Member 6 Transporter in HepG2 Cells: Effect on the Dynamics of Cell-Cell and Cell-Matrix Interactions. Int J Mol Sci 2023; 24:16391. [PMID: 38003580 PMCID: PMC10670978 DOI: 10.3390/ijms242216391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 11/12/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
There is growing evidence that various ATP-binding cassette (ABC) transporters contribute to the growth and development of tumors, but relatively little is known about how the ABC transporter family behaves in hepatocellular carcinoma (HCC), one of the most common cancers worldwide. Cellular model studies have shown that ABCC6, which belongs to the ABC subfamily C (ABCC), plays a role in the cytoskeleton rearrangement and migration of HepG2 hepatocarcinoma cells, thus highlighting its role in cancer biology. Deep knowledge on the molecular mechanisms underlying the observed results could provide therapeutic insights into the tumors in which ABCC6 is modulated. In this study, differential expression levels of mRNA transcripts between ABCC6-silenced HepG2 and control groups were measured, and subsequently, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed. Real-Time PCR and Western blot analyses confirmed bioinformatics; functional studies support the molecular mechanisms underlying the observed effects. The results provide valuable information on the dysregulation of fundamental cellular processes, such as the focal adhesion pathway, which allowed us to obtain detailed information on the active role that the down-regulation of ABCC6 could play in the biology of liver tumors, as it is involved not only in cell migration but also in cell adhesion and invasion.
Collapse
Affiliation(s)
- Ilenia Matera
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (I.M.); (R.M.); (V.A.); (M.A.C.M.)
| | - Rocchina Miglionico
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (I.M.); (R.M.); (V.A.); (M.A.C.M.)
| | - Vittorio Abruzzese
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (I.M.); (R.M.); (V.A.); (M.A.C.M.)
| | - Giovanna Marchese
- Genomix4Life Srl, 84081 Baronissi, Italy; (G.M.); (G.M.V.)
- Genome Research Center for Health—CRGS, 84081 Baronissi, Italy
| | | | | | - Faustino Bisaccia
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (I.M.); (R.M.); (V.A.); (M.A.C.M.)
| | - Angela Ostuni
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (I.M.); (R.M.); (V.A.); (M.A.C.M.)
| |
Collapse
|
8
|
Li NY, Vorrius B, Ge J, Qiao Z, Zhu S, Katarincic J, Chen Q. Matrilin-2 within a three-dimensional lysine-modified chitosan porous scaffold enhances Schwann cell migration and axonal outgrowth for peripheral nerve regeneration. Front Bioeng Biotechnol 2023; 11:1142610. [PMID: 37223659 PMCID: PMC10201561 DOI: 10.3389/fbioe.2023.1142610] [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: 01/11/2023] [Accepted: 04/20/2023] [Indexed: 05/25/2023] Open
Abstract
Background: Matrilin-2 is a key extracellular matrix protein involved in peripheral nerve regeneration. We sought to develop a biomimetic scaffold to enhance peripheral nerve regeneration by incorporating matrilin-2 within a chitosan-derived porous scaffold. We hypothesized that the use of such a novel biomaterial delivers microenvironmental cues to facilitate Schwann cell (SC) migration and enhance axonal outgrowth during peripheral nerve regeneration. Materials and Methods: The effect of matrilin-2 on SC migration was evaluated with agarose drop migration assay on matrilin-2 coated dishes. SC adhesion was determined with SCs cultured atop tissue culture dishes coated with matrilin-2. Various formulations of chitosan vs matrilin-2 in scaffold constructs were examined with scanning electron microscopy. The effect of the matrilin-2/chitosan scaffold on SC migration in the collagen conduits was determined by capillary migration assays. Neuronal adhesion and axonal outgrowth were evaluated with three-dimensional (3D) organotypic assay of dorsal root ganglions (DRG). DRG axonal outgrowth within the scaffolds was determined by immunofluorescence staining of neurofilaments. Results: Matrilin-2 induced SC migration and enhanced its adhesion. A formulation of 2% chitosan with matrilin-2 demonstrated an optimal 3D porous architecture for SC interaction. Matrilin-2/chitosan scaffold enabled SCs to migrate against gravity within conduits. Chemical modification of chitosan with lysine (K-chitosan) further improved DRG adhesion and axonal outgrowth than the matrilin-2/chitosan scaffold without lysine modification. Conclusion: We developed a matrilin-2/K-chitosan scaffold to mimic extracellular matrix cues and provide a porous matrix to enhance peripheral nerve regeneration. Taking advantage of matrilin-2's capability to stimulate SC migration and adhesion, we formulated a porous matrilin-2/chitosan scaffold to support axongal outgrowth. Chemical modification of chitosan with lysine further improved matrilin-2 bioactivity in the 3D scaffold. The 3D porous matrilin-2/K-chitosan scaffolds have high potential for enhancing nerve repair by stimulating SC migration, neuronal adhesion, and axonal outgrowth.
Collapse
Affiliation(s)
- Neill Y. Li
- Laboratory of Molecular Biology and Nanomedicine, Department of Orthopaedics, Warren Alpert Medical School of Brown University, Providence, RI, United States
- Department of Orthopaedics, Duke University School of Medicine, Durham, NC, United States
| | - Brandon Vorrius
- Laboratory of Molecular Biology and Nanomedicine, Department of Orthopaedics, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Jonathan Ge
- Laboratory of Molecular Biology and Nanomedicine, Department of Orthopaedics, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Zhen Qiao
- Laboratory of Molecular Biology and Nanomedicine, Department of Orthopaedics, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Shuang Zhu
- Laboratory of Molecular Biology and Nanomedicine, Department of Orthopaedics, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Julia Katarincic
- Laboratory of Molecular Biology and Nanomedicine, Department of Orthopaedics, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Qian Chen
- Laboratory of Molecular Biology and Nanomedicine, Department of Orthopaedics, Warren Alpert Medical School of Brown University, Providence, RI, United States
| |
Collapse
|
9
|
Rapp AE, Zaucke F. Cartilage extracellular matrix-derived matrikines in osteoarthritis. Am J Physiol Cell Physiol 2023; 324:C377-C394. [PMID: 36571440 DOI: 10.1152/ajpcell.00464.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Osteoarthritis (OA) is among the most frequent diseases of the musculoskeletal system. Degradation of cartilage extracellular matrix (ECM) is a hallmark of OA. During the degradation process, intact/full-length proteins and proteolytic fragments are released which then might induce different downstream responses via diverse receptors, therefore leading to different biological consequences. Collagen type II and the proteoglycan aggrecan are the most abundant components of the cartilage ECM. However, over the last decades, a large number of minor components have been identified and for some of those, a role in the manifold processes associated with OA has already been demonstrated. To date, there is still no therapy able to halt or cure OA. A better understanding of the matrikine landscape occurring with or even preceding obvious degenerative changes in joint tissues is needed and might help to identify molecules that could serve as biomarkers, druggable targets, or even be blueprints for disease modifying drug OA drugs. For this narrative review, we screened PubMed for relevant literature in the English language and summarized the current knowledge regarding the function of selected ECM molecules and the derived matrikines in the context of cartilage and OA.
Collapse
Affiliation(s)
- Anna E Rapp
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Frank Zaucke
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
10
|
Praus F, Künstner A, Sauer T, Kohl M, Kern K, Deichmann S, Végvári Á, Keck T, Busch H, Habermann JK, Gemoll T. Panomics reveals patient individuality as the major driver of colorectal cancer progression. J Transl Med 2023; 21:41. [PMID: 36691026 PMCID: PMC9869555 DOI: 10.1186/s12967-022-03855-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 12/26/2022] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most prevalent cancers, with over one million new cases per year. Overall, prognosis of CRC largely depends on the disease stage and metastatic status. As precision oncology for patients with CRC continues to improve, this study aimed to integrate genomic, transcriptomic, and proteomic analyses to identify significant differences in expression during CRC progression using a unique set of paired patient samples while considering tumour heterogeneity. METHODS We analysed fresh-frozen tissue samples prepared under strict cryogenic conditions of matched healthy colon mucosa, colorectal carcinoma, and liver metastasis from the same patients. Somatic mutations of known cancer-related genes were analysed using Illumina's TruSeq Amplicon Cancer Panel; the transcriptome was assessed comprehensively using Clariom D microarrays. The global proteome was evaluated by liquid chromatography-coupled mass spectrometry (LC‒MS/MS) and validated by two-dimensional difference in-gel electrophoresis. Subsequent unsupervised principal component clustering, statistical comparisons, and gene set enrichment analyses were calculated based on differential expression results. RESULTS Although panomics revealed low RNA and protein expression of CA1, CLCA1, MATN2, AHCYL2, and FCGBP in malignant tissues compared to healthy colon mucosa, no differentially expressed RNA or protein targets were detected between tumour and metastatic tissues. Subsequent intra-patient comparisons revealed highly specific expression differences (e.g., SRSF3, OLFM4, and CEACAM5) associated with patient-specific transcriptomes and proteomes. CONCLUSION Our research results highlight the importance of inter- and intra-tumour heterogeneity as well as individual, patient-paired evaluations for clinical studies. In addition to changes among groups reflecting CRC progression, we identified significant expression differences between normal colon mucosa, primary tumour, and liver metastasis samples from individuals, which might accelerate implementation of precision oncology in the future.
Collapse
Affiliation(s)
- Friederike Praus
- Section for Translational Surgical Oncology and Biobanking, Department of Surgery, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Axel Künstner
- Medical Systems Biology Group, Lübeck Institute Für Experimental Dermatology, University of Lübeck, Campus Lübeck, 23538, Lübeck, Germany
| | - Thorben Sauer
- Section for Translational Surgical Oncology and Biobanking, Department of Surgery, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Michael Kohl
- Section for Translational Surgical Oncology and Biobanking, Department of Surgery, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- Medical Systems Biology Group, Lübeck Institute Für Experimental Dermatology, University of Lübeck, Campus Lübeck, 23538, Lübeck, Germany
| | - Katharina Kern
- Section for Translational Surgical Oncology and Biobanking, Department of Surgery, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Steffen Deichmann
- Department of Surgery, University Hospital Schleswig-Holstein, Campus Lübeck, 23538, Lübeck, Germany
| | - Ákos Végvári
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
- Proteomics Biomedicum, Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Tobias Keck
- Department of Surgery, University Hospital Schleswig-Holstein, Campus Lübeck, 23538, Lübeck, Germany
| | - Hauke Busch
- Medical Systems Biology Group, Lübeck Institute Für Experimental Dermatology, University of Lübeck, Campus Lübeck, 23538, Lübeck, Germany
| | - Jens K Habermann
- Section for Translational Surgical Oncology and Biobanking, Department of Surgery, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- Department of Oncology Pathology, Karolinska Institutet, 171 64, Solna, Sweden
| | - Timo Gemoll
- Section for Translational Surgical Oncology and Biobanking, Department of Surgery, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany.
| |
Collapse
|
11
|
Saltarelli MA, Quarta A, Chiarelli F. Growth plate extracellular matrix defects and short stature in children. Ann Pediatr Endocrinol Metab 2022; 27:247-255. [PMID: 36567461 PMCID: PMC9816467 DOI: 10.6065/apem.2244120.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 06/29/2022] [Indexed: 12/27/2022] Open
Abstract
Many etiological factors causing short stature have already been identified in humans. In the last few years, the advent of new techniques for the detection of chromosomal and molecular abnormalities has made it possible to better identify patients with genetic causes of growth failure. Some of these factors directly affect the development and growth of the skeleton, since they damage the epiphyseal growth plate, where linear growth occurs, influencing chondrogenesis. In particular, defects in genes involved in the organization and function of the growth plate are responsible for several well-known conditions with short stature. These genes play a pivotal role in various mechanisms involving the extracellular matrix, intracellular signaling, paracrine signaling, endocrine signaling, and epigenetic regulation. In this review, we will discuss the genes involved in extracellular matrix disorders. The identification of genetic defects in linear growth failure is important for clinicians and researchers in order to improve the care of children affected by growth disorders.
Collapse
Affiliation(s)
| | - Alessia Quarta
- Department of Pediatrics, University of Chieti, Chieti, Italy
| | - Francesco Chiarelli
- Department of Pediatrics, University of Chieti, Chieti, Italy,Address for correspondence: Francesco Chiarelli Department of Pediatrics, University of Chieti, Via dei Vestini, 5 Chieti, I-66100, Italy
| |
Collapse
|
12
|
Boschiero C, Gao Y, Baldwin RL, Ma L, Li CJ, Liu GE. Differentially CTCF-Binding Sites in Cattle Rumen Tissue during Weaning. Int J Mol Sci 2022; 23:ijms23169070. [PMID: 36012336 PMCID: PMC9408924 DOI: 10.3390/ijms23169070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 11/17/2022] Open
Abstract
The weaning transition in calves is characterized by major structural changes such as an increase in the rumen capacity and surface area due to diet changes. Studies evaluating rumen development in calves are vital to identify genetic mechanisms affected by weaning. This study aimed to provide a genome-wide characterization of CTCF-binding sites and differentially CTCF-binding sites (DCBS) in rumen tissue during the weaning transition of four Holstein calves to uncover regulatory elements in rumen epithelial tissue using ChIP-seq. Our study generated 67,280 CTCF peaks for the before weaning (BW) and 39,891 for after weaning (AW). Then, 7401 DCBS were identified for the AW vs. BW comparison representing 0.15% of the cattle genome, comprising ~54% of induced DCBS and ~46% of repressed DCBS. Most of the induced and repressed DCBS were in distal intergenic regions, showing a potential role as insulators. Gene ontology enrichment revealed many shared GO terms for the induced and the repressed DCBS, mainly related to cellular migration, proliferation, growth, differentiation, cellular adhesion, digestive tract morphogenesis, and response to TGFβ. In addition, shared KEGG pathways were obtained for adherens junction and focal adhesion. Interestingly, other relevant KEGG pathways were observed for the induced DCBS like gastric acid secretion, salivary secretion, bacterial invasion of epithelial cells, apelin signaling, and mucin-type O-glycan biosynthesis. IPA analysis further revealed pathways with potential roles in rumen development during weaning, including TGFβ, Integrin-linked kinase, and Integrin signaling. When DCBS were further integrated with RNA-seq data, 36 putative target genes were identified for the repressed DCBS, including KRT84, COL9A2, MATN3, TSPAN1, and AJM1. This study successfully identified DCBS in cattle rumen tissue after weaning on a genome-wide scale and revealed several candidate target genes that may have a role in rumen development, such as TGFβ, integrins, keratins, and SMADs. The information generated in this preliminary study provides new insights into bovine genome regulation and chromatin landscape.
Collapse
Affiliation(s)
- Clarissa Boschiero
- Animal Genomics and Improvement Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD 20705, USA
| | - Yahui Gao
- Animal Genomics and Improvement Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD 20705, USA
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Ransom L. Baldwin
- Animal Genomics and Improvement Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD 20705, USA
| | - Li Ma
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Cong-jun Li
- Animal Genomics and Improvement Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD 20705, USA
- Correspondence: (C.-j.L.); (G.E.L.); Tel.: +1-301-504-7216 (C.-j.L.); +1-301-504-9843 (G.E.L.); Fax: +1-301-504-8414 (C.-j.L. & G.E.L.)
| | - George E. Liu
- Animal Genomics and Improvement Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD 20705, USA
- Correspondence: (C.-j.L.); (G.E.L.); Tel.: +1-301-504-7216 (C.-j.L.); +1-301-504-9843 (G.E.L.); Fax: +1-301-504-8414 (C.-j.L. & G.E.L.)
| |
Collapse
|
13
|
Lee SY, Lee JW. 3D Spheroid Cultures of Stem Cells and Exosome Applications for Cartilage Repair. LIFE (BASEL, SWITZERLAND) 2022; 12:life12070939. [PMID: 35888029 PMCID: PMC9317836 DOI: 10.3390/life12070939] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022]
Abstract
Cartilage is a connective tissue that constitutes the structure of the body and consists of chondrocytes that produce considerable collagenous extracellular matrix and plentiful ground substances, such as proteoglycan and elastin fibers. Self-repair is difficult when the cartilage is damaged because of insufficient blood supply, low cellularity, and limited progenitor cell numbers. Therefore, three-dimensional (3D) culture systems, including pellet culture, hanging droplets, liquid overlays, self-injury, and spinner culture, have attracted attention. In particular, 3D spheroid culture strategies can enhance the yield of exosome production of mesenchymal stem cells (MSCs) when compared to two-dimensional culture, and can improve cellular restorative function by enhancing the paracrine effects of MSCs. Exosomes are membrane-bound extracellular vesicles, which are intercellular communication systems that carry RNAs and proteins. Information transfer affects the phenotype of recipient cells. MSC-derived exosomes can facilitate cartilage repair by promoting chondrogenic differentiation and proliferation. In this article, we reviewed recent major advances in the application of 3D culture techniques, cartilage regeneration with stem cells using 3D spheroid culture system, the effect of exosomes on chondrogenic differentiation, and chondrogenic-specific markers related to stem cell derived exosomes. Furthermore, the utilization of MSC-derived exosomes to enhance chondrogenic differentiation for osteoarthritis is discussed. If more mechanistic studies at the molecular level are conducted, MSC-spheroid-derived exosomes will supply a better therapeutic option to improve osteoarthritis.
Collapse
Affiliation(s)
- Seung Yeon Lee
- Department of Molecular Medicine, College of Medicine, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Korea;
| | - Jin Woo Lee
- Department of Molecular Medicine, College of Medicine, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Korea;
- Department of Health Sciences and Technology, GAIHST, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Korea
- Correspondence: ; Tel.: +82-32-899-6516; Fax: +82-32-899-6039
| |
Collapse
|
14
|
Liu T, Li L, Cheng C, He B, Jiang T. Emerging prospects of protein/peptide-based nanoassemblies for drug delivery and vaccine development. NANO RESEARCH 2022; 15:7267-7285. [PMID: 35692441 PMCID: PMC9166156 DOI: 10.1007/s12274-022-4385-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 05/09/2023]
Abstract
Proteins have been widely used in the biomedical field because of their well-defined architecture, accurate molecular weight, excellent biocompatibility and biodegradability, and easy-to-functionalization. Inspired by the wisdom of nature, increasing proteins/peptides that possess self-assembling capabilities have been explored and designed to generate nanoassemblies with unique structure and function, including spatially organized conformation, passive and active targeting, stimuli-responsiveness, and high stability. These characteristics make protein/peptide-based nanoassembly an ideal platform for drug delivery and vaccine development. In this review, we focus on recent advances in subsistent protein/peptide-based nanoassemblies, including protein nanocages, virus-like particles, self-assemblable natural proteins, and self-assemblable artificial peptides. The origin and characteristics of various protein/peptide-based assemblies and their applications in drug delivery and vaccine development are summarized. In the end, the prospects and challenges are discussed for the further development of protein/peptide-based nanoassemblies.
Collapse
Affiliation(s)
- Taiyu Liu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816 China
| | - Lu Li
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816 China
| | - Cheng Cheng
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816 China
| | - Bingfang He
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816 China
| | - Tianyue Jiang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816 China
| |
Collapse
|
15
|
Valproic acid modulates collagen architecture in the postoperative conjunctival scar. J Mol Med (Berl) 2022; 100:947-961. [PMID: 35583819 DOI: 10.1007/s00109-021-02171-2] [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: 08/11/2021] [Revised: 11/10/2021] [Accepted: 11/29/2021] [Indexed: 10/18/2022]
Abstract
Valproic acid (VPA), widely used for the treatment of neurological disorders, has anti-fibrotic activity by reducing collagen production in the postoperative conjunctiva. In this study, we investigated the capacity of VPA to modulate the postoperative collagen architecture. Histochemical examination revealed that VPA treatment was associated with the formation of thinner collagen fibers in the postoperative days 7 and 14 scars. At the micrometer scale, measurements by quantitative multiphoton microscopy indicated that VPA reduced mean collagen fiber thickness by 1.25-fold. At the nanometer scale, collagen fibril thickness and diameter measured by transmission electron microscopy were decreased by 1.08- and 1.20-fold, respectively. Moreover, delicate filamentous structures in random aggregates or closely associated with collagen fibrils were frequently observed in VPA-treated tissue. At the molecular level, VPA reduced Col1a1 but induced Matn2, Matn3, and Matn4 in the postoperative day 7 conjunctival tissue. Elevation of matrilin protein expression induced by VPA was sustained till at least postoperative day 14. In primary conjunctival fibroblasts, Matn2 expression was resistant to both VPA and TGF-β2, Matn3 was sensitive to both VPA and TGF-β2 individually and synergistically, while Matn4 was modulable by VPA but not TGF-β2. MATN2, MATN3, and MATN4 localized in close association with COL1A1 in the postoperative conjunctiva. These data indicate that VPA has the capacity to reduce collagen fiber thickness and potentially collagen assembly, in association with matrilin upregulation. These properties suggest potential VPA application for the prevention of fibrotic progression in the postoperative conjunctiva. KEY MESSAGES: VPA reduces collagen fiber and fibril thickness in the postoperative scar. VPA disrupts collagen fiber assembly in conjunctival wound healing. VPA induces MATN2, MATN3, and MATN4 in the postoperative scar.
Collapse
|
16
|
Wang Z, Lee J, Breen C, Ruenger T, Xiong F, Goreshi R, Lu M, Iwamoto S. Peritumoral Matrilin-2 Staining May Be Useful in Distinguishing Basal Cell Carcinoma from Folliculocentric Basaloid Proliferation. J Cutan Pathol 2022; 49:543-548. [PMID: 35274748 DOI: 10.1111/cup.14222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/25/2022] [Accepted: 03/07/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Folliculocentric basaloid proliferation (FBP) is a benign and reactive proliferation, which can histopathologically mimic basal cell carcinomas (BCC). The incidental presence of FBP during the excision of a BCC can occasionally lead to excessive tissue removal. One distinguishing feature of BCCs is that they invade the stroma, whereas FBPs generally do not. METHODS Matrilin-2 is an extracellular matrix protein associated with tumor invasion, and we compared the expression of matrilin-2 in peritumoral cells of BCC and FBP. RESULTS We found increased matrilin-2 expression within the peritumoral stroma of 41 of 42 BCCs (97.7%), with strong expression in all (100%) cases of infiltrative and 21 of 25 (84%) nodular forms of BCC. We found no expression of peritumoral matrilin-2 in any of the seven cases of FBP. CONCLUSION Our results suggest that immunolabeling with the matrilin-2 antibody may help distinguish basal cell carcinomas from folliculocentric basaloid proliferations. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Zhengke Wang
- Division of Dermatology (Medicine) and Division of Dermatopathology (Pathology), Chartercare Roger Williams Medical Center, Providence, RI, United States
| | - Jennifer Lee
- Division of Dermatology (Medicine) and Division of Dermatopathology (Pathology), Chartercare Roger Williams Medical Center, Providence, RI, United States.,Boston University School of Medicine, Boston, MA, United States
| | - Catherine Breen
- Division of Dermatology (Medicine) and Division of Dermatopathology (Pathology), Chartercare Roger Williams Medical Center, Providence, RI, United States.,Boston University School of Medicine, Boston, MA, United States
| | - Thomas Ruenger
- Division of Dermatology (Medicine) and Division of Dermatopathology (Pathology), Chartercare Roger Williams Medical Center, Providence, RI, United States
| | - Fang Xiong
- Division of Dermatology (Medicine) and Division of Dermatopathology (Pathology), Chartercare Roger Williams Medical Center, Providence, RI, United States
| | - Renato Goreshi
- Division of Dermatology (Medicine) and Division of Dermatopathology (Pathology), Chartercare Roger Williams Medical Center, Providence, RI, United States
| | - Ming Lu
- Division of Dermatology (Medicine) and Division of Dermatopathology (Pathology), Chartercare Roger Williams Medical Center, Providence, RI, United States
| | - Satori Iwamoto
- Division of Dermatology (Medicine) and Division of Dermatopathology (Pathology), Chartercare Roger Williams Medical Center, Providence, RI, United States.,Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
17
|
Smith MM, Hayes AJ, Melrose J. Pentosan Polysulphate (PPS), a Semi-Synthetic Heparinoid DMOAD With Roles in Intervertebral Disc Repair Biology emulating The Stem Cell Instructive and Tissue Reparative Properties of Heparan Sulphate. Stem Cells Dev 2022; 31:406-430. [PMID: 35102748 DOI: 10.1089/scd.2022.0007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This review highlights the attributes of pentosan polysulphate (PPS) in the promotion of intervertebral disc (IVD) repair processes. PPS has been classified as a disease modifying osteoarthritic drug (DMOAD) and many studies have demonstrated its positive attributes in the countering of degenerative changes occurring in cartilaginous tissues during the development of osteoarthritis (OA). Degenerative changes in the IVD also involve inflammatory cytokines, degradative proteases and cell signalling pathways similar to those operative in the development of OA in articular cartilage. PPS acts as a heparan sulphate (HS) mimetic to effect its beneficial effects in cartilage. The IVD contains small cell membrane HS-proteoglycans (HSPGs) such as syndecan, and glypican and a large multifunctional HS/chondroitin sulphate (CS) hybrid proteoglycan (HSPG2/perlecan) that have important matrix stabilising properties and sequester, control and present growth factors from the FGF, VEGF, PDGF and BMP families to cellular receptors to promote cell proliferation, differentiation and matrix synthesis. HSPG2 also has chondrogenic properties and stimulates the synthesis of extracellular matrix (ECM) components, expansion of cartilaginous rudiments and has roles in matrix stabilisation and repair. Perlecan is a perinuclear and nuclear proteoglycan in IVD cells with roles in chromatin organisation and control of transcription factor activity, immunolocalises to stem cell niches in cartilage, promotes escape of stem cells from quiescent recycling, differentiation and attainment of pluripotency and migratory properties. These participate in tissue development and morphogenesis, ECM remodelling and repair. PPS also localises in the nucleus of stromal stem cells, promotes development of chondroprogenitor cell lineages, ECM synthesis and repair and discal repair by resident disc cells. The availability of recombinant perlecan and PPS offer new opportunities in repair biology. These multifunctional agents offer welcome new developments in repair strategies for the IVD.
Collapse
Affiliation(s)
- Margaret M Smith
- The University of Sydney Raymond Purves Bone and Joint Research Laboratories, 247198, St Leonards, New South Wales, Australia;
| | - Anthony J Hayes
- Cardiff School of Biosciences, University of Cardiff, UK, Bioimaging Unit, Cardiff, Wales, United Kingdom of Great Britain and Northern Ireland;
| | - James Melrose
- Kolling Institute, University of Sydney, Royal North Shore Hospital, Raymond Purves Lab, Sydney Medical School Northern, Level 10, Kolling Institute B6, Royal North Shore Hospital, St. Leonards, New South Wales, Australia, 2065.,University of New South Wales, 7800, Graduate School of Biomedical Engineering, University of NSW, Sydney, New South Wales, Australia, 2052;
| |
Collapse
|
18
|
Mechanical Cues: Bidirectional Reciprocity in the Extracellular Matrix Drives Mechano-Signalling in Articular Cartilage. Int J Mol Sci 2021; 22:ijms222413595. [PMID: 34948394 PMCID: PMC8707858 DOI: 10.3390/ijms222413595] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/08/2021] [Accepted: 12/15/2021] [Indexed: 12/29/2022] Open
Abstract
The composition and organisation of the extracellular matrix (ECM), particularly the pericellular matrix (PCM), in articular cartilage is critical to its biomechanical functionality; the presence of proteoglycans such as aggrecan, entrapped within a type II collagen fibrillar network, confers mechanical resilience underweight-bearing. Furthermore, components of the PCM including type VI collagen, perlecan, small leucine-rich proteoglycans—decorin and biglycan—and fibronectin facilitate the transduction of both biomechanical and biochemical signals to the residing chondrocytes, thereby regulating the process of mechanotransduction in cartilage. In this review, we summarise the literature reporting on the bidirectional reciprocity of the ECM in chondrocyte mechano-signalling and articular cartilage homeostasis. Specifically, we discuss studies that have characterised the response of articular cartilage to mechanical perturbations in the local tissue environment and how the magnitude or type of loading applied elicits cellular behaviours to effect change. In vivo, including transgenic approaches, and in vitro studies have illustrated how physiological loading maintains a homeostatic balance of anabolic and catabolic activities, involving the direct engagement of many PCM molecules in orchestrating this slow but consistent turnover of the cartilage matrix. Furthermore, we document studies characterising how abnormal, non-physiological loading including excessive loading or joint trauma negatively impacts matrix molecule biosynthesis and/or organisation, affecting PCM mechanical properties and reducing the tissue’s ability to withstand load. We present compelling evidence showing that reciprocal engagement of the cells with this altered ECM environment can thus impact tissue homeostasis and, if sustained, can result in cartilage degradation and onset of osteoarthritis pathology. Enhanced dysregulation of PCM/ECM turnover is partially driven by mechanically mediated proteolytic degradation of cartilage ECM components. This generates bioactive breakdown fragments such as fibronectin, biglycan and lumican fragments, which can subsequently activate or inhibit additional signalling pathways including those involved in inflammation. Finally, we discuss how bidirectionality within the ECM is critically important in enabling the chondrocytes to synthesise and release PCM/ECM molecules, growth factors, pro-inflammatory cytokines and proteolytic enzymes, under a specified load, to influence PCM/ECM composition and mechanical properties in cartilage health and disease.
Collapse
|
19
|
Jiang X, Wojtkiewicz M, Patwardhan C, Greer S, Kong Y, Kuss M, Huang X, Liao J, Lu Y, Dudley A, Gundry RL, Fuchs M, Streubel P, Duan B. The effects of maturation and aging on the rotator cuff tendon-to-bone interface. FASEB J 2021; 35:e22066. [PMID: 34822203 DOI: 10.1096/fj.202101484r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/29/2021] [Accepted: 11/10/2021] [Indexed: 12/19/2022]
Abstract
Rotator cuff tendon injuries often occur at the tendon-to-bone interface (i.e., enthesis) area, with a high prevalence for the elderly population, but the underlying reason for this phenomenon is still unknown. The objective of this study is to identify the histological, molecular, and biomechanical alterations of the rotator cuff enthesis with maturation and aging in a mouse model. Four different age groups of mice (newborn, young, adult, and old) were studied. Striking variations of the entheses were observed between the newborn and other matured groups, with collagen content, proteoglycan deposition, collagen fiber dispersion was significantly higher in the newborn group. The compositional and histological features of young, adult, and old groups did not show significant differences, except having increased proteoglycan deposition and thinner collagen fibers at the insertion sites in the old group. Nanoindentation testing showed that the old group had a smaller compressive modulus at the insertion site when compared with other groups. However, tensile mechanical testing reported that the old group demonstrated a significantly higher failure stress when compared with the young and adult groups. The proteomics analysis detected dramatic differences in protein content between newborn and young groups but minor changes among young, adult, and old groups. These results demonstrated: (1) the significant alterations of the enthesis composition and structure occur from the newborn to the young time period; (2) the increased risk of rotator cuff tendon injuries in the elderly population is not solely because of old age alone in the rodent model.
Collapse
Affiliation(s)
- Xiping Jiang
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Molecular Genetics and Cell Biology Program, Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Melinda Wojtkiewicz
- CardiOmics Program, Center for Heart and Vascular Research, Division of Cardiovascular Medicine, and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Chinmay Patwardhan
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Sydney Greer
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Molecular Genetics and Cell Biology Program, Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Yunfan Kong
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Mitchell Kuss
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Xi Huang
- Department of Electrical and Computer Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Yongfeng Lu
- Department of Electrical and Computer Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Andrew Dudley
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Molecular Genetics and Cell Biology Program, Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Rebekah L Gundry
- CardiOmics Program, Center for Heart and Vascular Research, Division of Cardiovascular Medicine, and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Matthias Fuchs
- Department of Physics and Astronomy, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Philipp Streubel
- Department of Orthopedic Surgery and Rehabilitation, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| |
Collapse
|
20
|
Ye L, Jin W. Identification of lncRNA-associated competing endogenous RNA networks for occurrence and prognosis of gastric carcinoma. J Clin Lab Anal 2021; 35:e24028. [PMID: 34704289 PMCID: PMC8649378 DOI: 10.1002/jcla.24028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 12/24/2022] Open
Abstract
Background Gastric cancer (GC) is one of the common digestive malignancies worldwide and causes a severe public health issue. So far, the underlying mechanisms of GC are largely unclear. Thus, we aim to identify the long non‐coding RNA (lncRNA)‐associated competing endogenous RNA (ceRNA) for GC. Methods TCGA database was downloaded and used for the identification of differentially expressed (DE) lncRNAs, miRNAs, and mRNAs, respectively. Then, the ceRNA network was constructed via multiple online datasets and approaches. In addition, various in vitro assays were carried out to validate the effect of certain hub lncRNAs. Results We constructed a ceRNA network, including 76 lncRNAs, 18 miRNAs, and 159 mRNAs, which involved multiple critical pathways. Next, univariate and multivariate analysis demonstrated 11 lncRNAs, including LINC02731, MIR99AHG, INHBA‐AS1, CCDC144NL‐AS1, VLDLR‐AS1, LIFR‐AS1, A2M‐AS1, LINC01537, and LINC00702, and were associated with OS, and nine of those lncRNAs were considered as hub lncRNAs involved in the sub‐ceRNA network. The in vitro assay indicated two lncRNAs, INHBA‐AS1 and CCDC144NL‐AS1, which were positively related to the GC aggressive features, including proliferation, invasion, and migration. Conclusions We identified nine hub lncRNAs and the associated ceRNA network related to the prognosis of GC, and then validated two out of them as promising oncogenes in GC.
Collapse
Affiliation(s)
- Lianmin Ye
- Department of Intensive Care, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wumin Jin
- Department of Reproductive Medicine Centre, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
21
|
Semba RD, Zhang P, Dufresne C, Gao T, Al-Jadaan I, Craven ER, Qian J, Edward DP, Mahale A. Primary angle closure glaucoma is characterized by altered extracellular matrix homeostasis in the iris. Proteomics Clin Appl 2021; 15:e2000094. [PMID: 34240827 DOI: 10.1002/prca.202000094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 06/19/2021] [Accepted: 07/06/2021] [Indexed: 11/10/2022]
Abstract
PURPOSE To characterize the proteome of the iris in primary angle closure glaucoma (PACG). EXPERIMENTAL DESIGN In this cross-sectional study, iris samples were obtained from surgical iridectomy of 48 adults with PACG and five normal controls. Peptides from iris were analysed using liquid chromatography-tandem mass spectrometry on an Orbitrap Q Exactive Plus mass spectrometer. Verification of proteins of interest was conducted using selected reaction monitoring on a triple quadrupole mass spectrometer. The main outcome was proteins with a log2 two-fold difference in expression in iris between PACG and controls. RESULTS There were 3,446 non-redundant proteins identified in human iris, of which 416 proteins were upregulated and 251 proteins were downregulated in PACG compared with controls. Thirty-two upregulated proteins were either components of the extracellular matrix (ECM) (fibrillar collagens, EMILIN-2, fibrinogen, fibronectin, matrilin-2), matricellular proteins (thrombospondin-1), proteins involved in cell-matrix interactions (integrins, laminin, histidine-rich glycoprotein, paxillin), or protease inhibitors known to modulate ECM turnover (α-2 macroglobulin, tissue factor pathway inhibitor 2, papilin). Two giant proteins, titin and obscurin, were up- and down-regulated, respectively, in the iris in PACG compared with controls. CONCLUSIONS AND CLINICAL RELEVANCE This proteomic study shows that ECM composition and homeostasis are altered in the iris in PACG.
Collapse
Affiliation(s)
- Richard D Semba
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Pingbo Zhang
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Tianshun Gao
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Earl R Craven
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jiang Qian
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Deepak P Edward
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,King Khaled Eye Specialist Hospital, Riyadh, Saudi Arabia
| | - Alka Mahale
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,King Khaled Eye Specialist Hospital, Riyadh, Saudi Arabia
| |
Collapse
|
22
|
Muttigi MS, Kim BJ, Choi B, Han I, Park H, Lee SH. Matrilin-3-Primed Adipose-Derived Mesenchymal Stromal Cell Spheroids Prevent Mesenchymal Stromal-Cell-Derived Chondrocyte Hypertrophy. Int J Mol Sci 2020; 21:ijms21238911. [PMID: 33255398 PMCID: PMC7727796 DOI: 10.3390/ijms21238911] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/20/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023] Open
Abstract
Adipose-derived mesenchymal stromal cells (Ad-MSCs) are a promising tool for articular cartilage repair and regeneration. However, the terminal hypertrophic differentiation of Ad-MSC-derived cartilage is a critical barrier during hyaline cartilage regeneration. In this study, we investigated the role of matrilin-3 in preventing Ad-MSC-derived chondrocyte hypertrophy in vitro and in an osteoarthritis (OA) destabilization of the medial meniscus (DMM) model. Methacrylated hyaluron (MAHA) (1%) was used to encapsulate and make scaffolds containing Ad-MSCs and matrilin-3. Subsequently, the encapsulated cells in the scaffolds were differentiated in chondrogenic medium (TGF-β, 1-14 days) and thyroid hormone hypertrophic medium (T3, 15-28 days). The presence of matrilin-3 with Ad-MSCs in the MAHA scaffold significantly increased the chondrogenic marker and decreased the hypertrophy marker mRNA and protein expression. Furthermore, matrilin-3 significantly modified the expression of TGF-β2, BMP-2, and BMP-4. Next, we prepared the OA model and transplanted Ad-MSCs primed with matrilin-3, either as a single-cell suspension or in spheroid form. Safranin-O staining and the OA score suggested that the regenerated cartilage morphology in the matrilin-3-primed Ad-MSC spheroids was similar to the positive control. Furthermore, matrilin-3-primed Ad-MSC spheroids prevented subchondral bone sclerosis in the mouse model. Here, we show that matrilin-3 plays a major role in modulating Ad-MSCs' therapeutic effect on cartilage regeneration and hypertrophy suppression.
Collapse
Affiliation(s)
| | - Byoung Ju Kim
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea;
| | - Bogyu Choi
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, 13488 Seongnam, Korea;
| | - Inbo Han
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Korea;
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul 06911, Korea;
- Correspondence: (H.P.); (S.-H.L.); Tel.: +82-2-820-5804 (H.P.); +82-31-961-5153 (S.-H.L.); Fax: +82-2-813-8159 (H.P.); +82-31-961-5108 (S.-H.L.)
| | - Soo-Hong Lee
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea;
- Correspondence: (H.P.); (S.-H.L.); Tel.: +82-2-820-5804 (H.P.); +82-31-961-5153 (S.-H.L.); Fax: +82-2-813-8159 (H.P.); +82-31-961-5108 (S.-H.L.)
| |
Collapse
|
23
|
Westermann LM, Fleischhauer L, Vogel J, Jenei-Lanzl Z, Ludwig NF, Schau L, Morellini F, Baranowsky A, Yorgan TA, Di Lorenzo G, Schweizer M, de Souza Pinheiro B, Guarany NR, Sperb-Ludwig F, Visioli F, Oliveira Silva T, Soul J, Hendrickx G, Wiegert JS, Schwartz IVD, Clausen-Schaumann H, Zaucke F, Schinke T, Pohl S, Danyukova T. Imbalanced cellular metabolism compromises cartilage homeostasis and joint function in a mouse model of mucolipidosis type III gamma. Dis Model Mech 2020; 13:dmm046425. [PMID: 33023972 PMCID: PMC7687858 DOI: 10.1242/dmm.046425] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/15/2020] [Indexed: 11/23/2022] Open
Abstract
Mucolipidosis type III (MLIII) gamma is a rare inherited lysosomal storage disorder caused by mutations in GNPTG encoding the γ-subunit of GlcNAc-1-phosphotransferase, the key enzyme ensuring proper intracellular location of multiple lysosomal enzymes. Patients with MLIII gamma typically present with osteoarthritis and joint stiffness, suggesting cartilage involvement. Using Gnptg knockout (Gnptgko ) mice as a model of the human disease, we showed that missorting of a number of lysosomal enzymes is associated with intracellular accumulation of chondroitin sulfate in Gnptgko chondrocytes and their impaired differentiation, as well as with altered microstructure of the cartilage extracellular matrix (ECM). We also demonstrated distinct functional and structural properties of the Achilles tendons isolated from Gnptgko and Gnptab knock-in (Gnptabki ) mice, the latter displaying a more severe phenotype resembling mucolipidosis type II (MLII) in humans. Together with comparative analyses of joint mobility in MLII and MLIII patients, these findings provide a basis for better understanding of the molecular reasons leading to joint pathology in these patients. Our data suggest that lack of GlcNAc-1-phosphotransferase activity due to defects in the γ-subunit causes structural changes within the ECM of connective and mechanosensitive tissues, such as cartilage and tendon, and eventually results in functional joint abnormalities typically observed in MLIII gamma patients. This idea was supported by a deficit of the limb motor function in Gnptgko mice challenged on a rotarod under fatigue-associated conditions, suggesting that the impaired motor performance of Gnptgko mice was caused by fatigue and/or pain at the joint.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Lena Marie Westermann
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lutz Fleischhauer
- Laboratory of Experimental Surgery and Regenerative Medicine, Clinic for General Trauma and Reconstructive Surgery, Ludwig-Maximilians University, 80336 Munich, Germany
- Center for Applied Tissue Engineering and Regenerative Medicine (Canter), University of Applied Sciences, 80533 Munich, Germany
| | - Jonas Vogel
- Center for Applied Tissue Engineering and Regenerative Medicine (Canter), University of Applied Sciences, 80533 Munich, Germany
| | - Zsuzsa Jenei-Lanzl
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopedic University Hospital Friedrichsheim gGmbH, 60528 Frankfurt/Main, Germany
| | - Nataniel Floriano Ludwig
- Post-Graduate Program in Genetics and Molecular Biology, Federal University of Rio Grande do Sul, 90040-060 Porto Alegre, Brazil
| | - Lynn Schau
- RG Behavioral Biology, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Fabio Morellini
- RG Behavioral Biology, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Anke Baranowsky
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Timur A Yorgan
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Giorgia Di Lorenzo
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Michaela Schweizer
- Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Bruna de Souza Pinheiro
- Department of Genetics, Federal University of Rio Grande do Sul, 90040-060 Porto Alegre, Brazil
| | - Nicole Ruas Guarany
- Occupational Therapy Faculty, Federal University of Pelotas, 96010-610 Pelotas, Brazil
| | - Fernanda Sperb-Ludwig
- Department of Genetics, Federal University of Rio Grande do Sul, 90040-060 Porto Alegre, Brazil
| | - Fernanda Visioli
- Pathology Department, Federal University of Rio Grande do Sul, 90040-060 Porto Alegre, Brazil
| | - Thiago Oliveira Silva
- Post-Graduate Program in Medicine: Medical Sciences, Federal University of Rio Grande do Sul, 90040-060 Porto Alegre, Brazil
| | - Jamie Soul
- Skeletal Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Gretl Hendrickx
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - J Simon Wiegert
- RG Synaptic Wiring and Information Processing, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Ida V D Schwartz
- Department of Genetics, Federal University of Rio Grande do Sul, 90040-060 Porto Alegre, Brazil
- Post-Graduate Program in Medicine: Medical Sciences, Federal University of Rio Grande do Sul, 90040-060 Porto Alegre, Brazil
| | - Hauke Clausen-Schaumann
- Center for Applied Tissue Engineering and Regenerative Medicine (Canter), University of Applied Sciences, 80533 Munich, Germany
| | - Frank Zaucke
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopedic University Hospital Friedrichsheim gGmbH, 60528 Frankfurt/Main, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Sandra Pohl
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Tatyana Danyukova
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
24
|
Muttigi MS, Kim BJ, Kumar H, Park S, Choi UY, Han I, Park H, Lee SH. Efficacy of matrilin-3-primed adipose-derived mesenchymal stem cell spheroids in a rabbit model of disc degeneration. Stem Cell Res Ther 2020; 11:363. [PMID: 32831130 PMCID: PMC7444036 DOI: 10.1186/s13287-020-01862-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 05/27/2020] [Accepted: 07/28/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Chronic low back pain is a prevalent disability, often caused by intervertebral disc (IVD) degeneration. Mesenchymal stem cell (MSC) therapy could be a safe and feasible option for repairing the degenerated disc. However, for successful translation to the clinic, various challenges need to be overcome including unwanted adverse effects due to acidic pH, hypoxia, and limited nutrition. Matrilin-3 is an essential extracellular matrix (ECM) component during cartilage development and ossification and exerts chondrocyte protective effects. METHODS This study evaluated the effects of matrilin-3-primed adipose-derived MSCs (Ad-MSCs) on the repair of the degenerated disc in vitro and in vivo. We determined the optimal priming concentration and duration and developed an optimal protocol for Ad-MSC spheroid generation. RESULTS Priming with 10 ng/ml matrilin-3 for 5 days resulted in the highest mRNA expression of type 2 collagen and aggrecan in vitro. Furthermore, Ad-MSC spheroids with a density of 250 cells/microwell showed the increased secretion of favorable growth factors such as transforming growth factor beta (TGF-β1), TGF-β2, interleukin-10 (IL-10), granulocyte colony-stimulating factor (G-CSF), and matrix metalloproteinase 1 (MMP1) and decreased secretion of hypertrophic ECM components. In addition, matrilin-3-primed Ad-MSC spheroid implantation was associated with optimal repair in a rabbit model. CONCLUSION Our results suggest that priming MSCs with matrilin-3 and spheroid formation could be an effective strategy to overcome the challenges associated with the use of MSCs for the treatment of IVD degeneration.
Collapse
Affiliation(s)
- Manjunatha S Muttigi
- School of Integrative Engineering, Chung-Ang University, Seoul, 06911, South Korea
| | - Byoung Ju Kim
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul, 04620, South Korea
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, 382010, India
| | - Sunghyun Park
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul, 04620, South Korea
- Department of Biomedical Science, CHA University, Seongnam-si, 13488, South Korea
| | - Un Yong Choi
- Department of Neurosurgery, School of Medicine, CHA Bundang Medical Center, CHA University, Seongnam-si, 13496, South Korea
| | - Inbo Han
- Department of Neurosurgery, School of Medicine, CHA Bundang Medical Center, CHA University, Seongnam-si, 13496, South Korea.
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul, 06911, South Korea.
| | - Soo-Hong Lee
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul, 04620, South Korea.
| |
Collapse
|
25
|
Zhang X, Zhou Q, Li X, Zou W, Hu X. Integrating omics and traditional analyses to profile the synergistic toxicity of graphene oxide and triphenyl phosphate. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 263:114473. [PMID: 33618456 DOI: 10.1016/j.envpol.2020.114473] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 03/02/2020] [Accepted: 03/25/2020] [Indexed: 06/12/2023]
Abstract
The increasing production and applications of graphene oxide (GO, a novel carbon nanomaterial) have raised numerous environmental concerns regarding its ecological risks. Triphenyl phosphate (TPhP) disperses in water and poses an increasing hazard to the ecosystem and human health. It is critical to study the environmental responses and molecular mechanisms of GO and TPhP together to assess both chemicals; however, this information is lacking. The present work revealed that GO promoted the bioaccumulation of TPhP in zebrafish larvae by 5.0%-24.3%. The TPhP-induced growth inhibition of embryos (malformation, mortality, heartbeat, and spontaneous movement) at environmentally relevant concentrations was significantly amplified by GO, and these results were supported by the downregulated levels of genes and proteins associated with cytoskeletal construction and cartilage and eye development. TPhP induced negligible alterations in the genes or proteins involved in oxidative stress and apoptosis, but those related proteins were all upregulated by GO. GO and TPhP coexposure activated the mTOR signaling pathway and subsequently promoted apoptosis in zebrafish by potentiating the oxidative stress induced by TPhP, presenting synergistic toxicity. These findings highlight the potential risks and specific molecular mechanisms of combining emerging carbon nanomaterials with coexisting organic contaminants.
Collapse
Affiliation(s)
- Xingli Zhang
- School of Environment, Key Laboratory for Yellow River and Huai River Water Environment and Pollution Control, Ministry of Education, Henan Key Laboratory for Environmental Pollution Control, International Joint Laboratory on Key Techniques in Water Treatment, Henan Normal University, Xinxiang, 453007, China
| | - Qixing Zhou
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education)/Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin, 300350, China.
| | - Xinyu Li
- School of Environment, Key Laboratory for Yellow River and Huai River Water Environment and Pollution Control, Ministry of Education, Henan Key Laboratory for Environmental Pollution Control, International Joint Laboratory on Key Techniques in Water Treatment, Henan Normal University, Xinxiang, 453007, China
| | - Wei Zou
- School of Environment, Key Laboratory for Yellow River and Huai River Water Environment and Pollution Control, Ministry of Education, Henan Key Laboratory for Environmental Pollution Control, International Joint Laboratory on Key Techniques in Water Treatment, Henan Normal University, Xinxiang, 453007, China.
| | - Xiangang Hu
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education)/Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin, 300350, China
| |
Collapse
|
26
|
Wilhelm D, Kempf H, Bianchi A, Vincourt JB. ATDC5 cells as a model of cartilage extracellular matrix neosynthesis, maturation and assembly. J Proteomics 2020; 219:103718. [PMID: 32097723 DOI: 10.1016/j.jprot.2020.103718] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/05/2020] [Accepted: 02/19/2020] [Indexed: 01/03/2023]
Abstract
Fibrillar collagens and proteoglycans (PGs) are quantitatively the major constituents of extracellular matrices (ECM). They carry numerous crucial post-translational modifications (PTMs) that tune the resulting biomechanical properties of the corresponding tissues. The mechanisms determining these PTMs remain largely unknown, notably because available established cell lines do not recapitulate much of the complexity of the machineries involved. ATDC5 cells are a model of chondrogenesis widely used for decades, but it remains described mostly at histological and transcriptional levels. Here, we asked to what extent this model recapitulates the events of ECM synthesis and processing occurring in cartilage. Insulin-stimulated ATDC5 cells exhibit up- or down-regulation of more than one-hundred proteins, including a number of known participants in chondrogenesis and major markers thereof. However, they also lack several ECM components considered of significant, yet more subtle, function in cartilage. Still, they assemble the large PG aggrecan and type II collagen, both carrying most of their in vivo PTMs, into an ECM. Remarkably, collagen crosslinking is fully lysyl oxidase (LOX)-dependent. The ATDC5 model recapitulates critical aspects of the cartilage ECM-processing machinery and should be useful to decipher the mechanisms involved. Proteomics data are available via ProteomeXchange with identifier PXD014121. SIGNIFICANCE: The present work provides the first proteome characterization of the ATDC5 chondrogenesis model, which has been used for decades in the field of cartilage biology. The results demonstrate the up- and down-regulation of more than one hundred proteins. Overall, specific drawbacks of the model are pointed out, that will be important to take into consideration for future studies. However, major cartilage components are massively assembled into an extracellular matrix and carry most of their post-translational modifications occurring in cartilage tissue. Unlike other available established cell lines, the ATDC5 model recapitulates major aspects of cartilage biosynthesis and should be useful in investigating the mechanisms that regulate collagen maturation events.
Collapse
Affiliation(s)
- Dafné Wilhelm
- UMR 7365 CNRS-UL IMoPA, Vandoeuvre-lès-Nancy, France
| | - Hervé Kempf
- UMR 7365 CNRS-UL IMoPA, Vandoeuvre-lès-Nancy, France
| | | | - Jean-Baptiste Vincourt
- UMR 7365 CNRS-UL IMoPA, Vandoeuvre-lès-Nancy, France; Proteomics core facility of UMS 2008 UL-CNRS-INSERM IBSLor, Vandoeuvre-lès-Nancy, France.
| |
Collapse
|
27
|
The E, Yao Q, Zhang P, Zhai Y, Ao L, Fullerton DA, Meng X. Mechanistic Roles of Matrilin-2 and Klotho in Modulating the Inflammatory Activity of Human Aortic Valve Cells. Cells 2020; 9:cells9020385. [PMID: 32046115 PMCID: PMC7072362 DOI: 10.3390/cells9020385] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 01/30/2020] [Accepted: 02/05/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Calcific aortic valve disease (CAVD) is a chronic inflammatory disease. Soluble extracellular matrix (ECM) proteins can act as damage-associated molecular patterns and may induce valvular inflammation. Matrilin-2 is an ECM protein and has been found to elevate the pro-osteogenic activity in human aortic valve interstitial cells (AVICs). Klotho, an anti-aging protein, appears to have anti-inflammatory properties. The effect of matrilin-2 and Klotho on AVIC inflammatory responses remains unclear. METHODS AND RESULTS Isolated human AVICs were exposed to matrilin-2. Soluble matrilin-2 induced the production of ICAM-1, MCP-1, and IL-6. It also induced protein kinase R (PKR) activation via Toll-like receptor (TLR) 2 and 4. Pretreatment with PKR inhibitors inhibited NF-κB activation and inflammatory mediator production induced by matrilin-2. Further, recombinant Klotho suppressed PKR and NF-κB activation and markedly reduced the production of inflammatory mediators in human AVICs exposed to matrilin-2. CONCLUSIONS This study revealed that soluble matrilin-2 upregulates AVIC inflammatory activity via activation of the TLR-PKR-NF-κB pathway and that Klotho is potent to suppress AVIC inflammatory responses to a soluble ECM protein through inhibiting PKR. These novel findings indicate that soluble matrilin-2 may accelerate the progression of CAVD by inducing valvular inflammation and that Klotho has the potential to suppress valvular inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xianzhong Meng
- Correspondence: ; Tel.: +1-303-724-6303; Fax: +1-303-724-6330
| |
Collapse
|
28
|
García-Alvarado FJ, Delgado-Aguirre HA, Rosales-González M, González-Martínez MDR, Ruiz-Flores P, González-Galarza FF, Arellano Perez Vertti RD. Analysis of Polymorphisms in the MATN3 and DOT1L Genes and CTX-II Urinary Levels in Patients with Knee Osteoarthritis in a Northeast Mexican-Mestizo Population. Genet Test Mol Biomarkers 2020; 24:105-111. [DOI: 10.1089/gtmb.2019.0179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Francisco J. García-Alvarado
- Departamento de Investigación, Universidad Juárez del Estado de Durango, Facultad de Ciencias de la Salud, Gómez Palacio Durango, México
| | - Héctor A. Delgado-Aguirre
- Departamento de Trasplantes, Instituto Mexicano del Seguro Social Hospital de Especialidades 71, Torreón, México
| | - Manuel Rosales-González
- Departamento de Investigación, Universidad Juárez del Estado de Durango, Facultad de Ciencias de la Salud, Gómez Palacio Durango, México
| | | | - Pablo Ruiz-Flores
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad Autónoma de Coahuila, Torreón, México
| | - Faviel F. González-Galarza
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad Autónoma de Coahuila, Torreón, México
| | | |
Collapse
|
29
|
A case of localized tracheobronchial relapsing polychondritis with positive matrilin-1 staining. BMC Rheumatol 2020; 4:1. [PMID: 32016169 PMCID: PMC6988282 DOI: 10.1186/s41927-019-0103-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 10/30/2019] [Indexed: 11/24/2022] Open
Abstract
Background Relapsing polychondritis (RPC) is a rare progressive autoimmune disease characterized by inflammation in the cartilage of multiple organs. Tracheobronchial involvement appears in nearly half of RPC patients during the course of their disease and represents the main cause of death. Localized tracheobronchial RPC is much rarer, and the pathogenesis remains unclear. Matrilin-1 is a non-collagenous cartilage matrix protein and has been suggested to be a potent autoantigen that induces the airway disease of RPC in animal models. However, the expression of matrilin-1 in tracheobronchial tissue in human remains unclear. Therefore, we examined the expression of matrilin-1 in the tracheal and auricular tissues in a localized tracheobronchial RPC patient. Case presentation A 62-year-old man with systemic sclerosis presented with cough and dyspnea on exertion. The lung function test showed an expiratory flow limitation and chest computed tomography showed diffuse thickness from the trachea to the bronchiole. No other tests showed abnormal findings. To evaluate further, bronchoscopy was performed and endobronchial ultrasonography showed thickness in the fourth-marginal echo layer suggesting inflammation of the cartilage. However, the tracheal biopsy showed no specific findings. The subsequent surgical tracheal biopsies showed inflammatory cell infiltration with destruction of the cartilage. Neither auricular nor nasal deformity, except for a tracheobronchial lesion, was detected. Biopsy from the left auricular cartilage also did not show any inflammatory changes. Finally, we diagnosed the patient with localized tracheobronchial RPC. To address the hypothesis that autoimmunity against matrilin-1 is involved in the pathogenesis of localized tracheobronchial RPC, we evaluated the expression level of matrilin-1 in a tracheal and auricular specimen from this patient. Immunohistochemical staining with anti-matrilin-1 antibody showed matrilin-1 in the tracheal but not in the auricular cartilage. Conclusions We first demonstrated the expression of matrilin-1 in tracheal but not in auricular cartilage in a localized tracheobronchial RPC patient. This result supports the possibility that matrilin-1 is involved in the pathogenesis of localized tracheobronchial RPC. However, this is only one case report and further observations will be needed to confirm this result.
Collapse
|
30
|
Li P, Fleischhauer L, Nicolae C, Prein C, Farkas Z, Saller MM, Prall WC, Wagener R, Heilig J, Niehoff A, Clausen-Schaumann H, Alberton P, Aszodi A. Mice Lacking the Matrilin Family of Extracellular Matrix Proteins Develop Mild Skeletal Abnormalities and Are Susceptible to Age-Associated Osteoarthritis. Int J Mol Sci 2020; 21:ijms21020666. [PMID: 31963938 PMCID: PMC7013758 DOI: 10.3390/ijms21020666] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/10/2020] [Accepted: 01/15/2020] [Indexed: 12/14/2022] Open
Abstract
Matrilins (MATN1, MATN2, MATN3 and MATN4) are adaptor proteins of the cartilage extracellular matrix (ECM), which bridge the collagen II and proteoglycan networks. In humans, dominant-negative mutations in MATN3 lead to various forms of mild chondrodysplasias. However, single or double matrilin knockout mice generated previously in our laboratory do not show an overt skeletal phenotype, suggesting compensation among the matrilin family members. The aim of our study was to establish a mouse line, which lacks all four matrilins and analyze the consequence of matrilin deficiency on endochondral bone formation and cartilage function. Matn1-4−/− mice were viable and fertile, and showed a lumbosacral transition phenotype characterized by the sacralization of the sixth lumbar vertebra. The development of the appendicular skeleton, the structure of the growth plate, chondrocyte differentiation, proliferation, and survival were normal in mutant mice. Biochemical analysis of knee cartilage demonstrated moderate alterations in the extractability of the binding partners of matrilins in Matn1-4−/− mice. Atomic force microscopy (AFM) revealed comparable compressive stiffness but higher collagen fiber diameters in the growth plate cartilage of quadruple mutant compared to wild-type mice. Importantly, Matn1-4−/− mice developed more severe spontaneous osteoarthritis at the age of 18 months, which was accompanied by changes in the biomechanical properties of the articular cartilage. Interestingly, Matn4−/− mice also developed age-associated osteoarthritis suggesting a crucial role of MATN4 in maintaining the stability of the articular cartilage. Collectively, our data provide evidence that matrilins are important to protect articular cartilage from deterioration and are involved in the specification of the vertebral column.
Collapse
Affiliation(s)
- Ping Li
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Ludwig-Maximilians-University, 80336 Munich, Germany; (P.L.); (L.F.); (C.P.); (Z.F.); (M.M.S.); (W.C.P.); (P.A.)
| | - Lutz Fleischhauer
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Ludwig-Maximilians-University, 80336 Munich, Germany; (P.L.); (L.F.); (C.P.); (Z.F.); (M.M.S.); (W.C.P.); (P.A.)
- Center for Applied Tissue Engineering and Regenerative Medicine, Munich University of Applied Sciences, 80533 Munich, Germany;
- Center for NanoScience, Ludwig-Maximilians University Munich, 80799 Munich, Germany
| | - Claudia Nicolae
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany;
| | - Carina Prein
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Ludwig-Maximilians-University, 80336 Munich, Germany; (P.L.); (L.F.); (C.P.); (Z.F.); (M.M.S.); (W.C.P.); (P.A.)
- Center for Applied Tissue Engineering and Regenerative Medicine, Munich University of Applied Sciences, 80533 Munich, Germany;
| | - Zsuzsanna Farkas
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Ludwig-Maximilians-University, 80336 Munich, Germany; (P.L.); (L.F.); (C.P.); (Z.F.); (M.M.S.); (W.C.P.); (P.A.)
| | - Maximilian Michael Saller
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Ludwig-Maximilians-University, 80336 Munich, Germany; (P.L.); (L.F.); (C.P.); (Z.F.); (M.M.S.); (W.C.P.); (P.A.)
| | - Wolf Christian Prall
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Ludwig-Maximilians-University, 80336 Munich, Germany; (P.L.); (L.F.); (C.P.); (Z.F.); (M.M.S.); (W.C.P.); (P.A.)
| | - Raimund Wagener
- Center for Molecular Medicine, University of Cologne, 50923 Cologne, Germany;
- Center for Biochemistry, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany;
| | - Juliane Heilig
- Center for Biochemistry, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany;
- Cologne Center for Musculoskeletal Biomechanics, Faculty of Medicine and University Hospital of Cologne, 50931 Cologne, Germany;
| | - Anja Niehoff
- Cologne Center for Musculoskeletal Biomechanics, Faculty of Medicine and University Hospital of Cologne, 50931 Cologne, Germany;
- Institute of Biomechanics and Orthopaedics, German Sport University Cologne, 50933 Cologne, Germany
| | - Hauke Clausen-Schaumann
- Center for Applied Tissue Engineering and Regenerative Medicine, Munich University of Applied Sciences, 80533 Munich, Germany;
- Center for NanoScience, Ludwig-Maximilians University Munich, 80799 Munich, Germany
| | - Paolo Alberton
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Ludwig-Maximilians-University, 80336 Munich, Germany; (P.L.); (L.F.); (C.P.); (Z.F.); (M.M.S.); (W.C.P.); (P.A.)
| | - Attila Aszodi
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Ludwig-Maximilians-University, 80336 Munich, Germany; (P.L.); (L.F.); (C.P.); (Z.F.); (M.M.S.); (W.C.P.); (P.A.)
- Center for Applied Tissue Engineering and Regenerative Medicine, Munich University of Applied Sciences, 80533 Munich, Germany;
- Correspondence: ; Tel.: +49-89-4400-55481
| |
Collapse
|
31
|
Vitamin K effects in human health: new insights beyond bone and cardiovascular health. J Nephrol 2019; 33:239-249. [DOI: 10.1007/s40620-019-00685-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 12/02/2019] [Indexed: 12/31/2022]
|
32
|
Baumgartner M, Drake K, Kanadia RN. An Integrated Model of Minor Intron Emergence and Conservation. Front Genet 2019; 10:1113. [PMID: 31798628 PMCID: PMC6865273 DOI: 10.3389/fgene.2019.01113] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 10/16/2019] [Indexed: 12/12/2022] Open
Abstract
Minor introns constitute <0.5% of the introns in the human genome and have remained an enigma since their discovery. These introns are removed by a distinct splicing complex, the minor spliceosome. Both are ancient, tracing back to the last eukaryotic common ancestor (LECA), which is reflected by minor intron enrichment in specific gene families, such as the mitogen activated-protein kinase kinases, voltage-gated sodium and calcium ion channels, and E2F transcription factors. Most minor introns occur as single introns in genes with predominantly major introns. Due to this organization, minor intron-containing gene (MIG) expression requires the coordinated action of two spliceosomes, which increases the probability of missplicing. Thus, one would expect loss of minor introns via purifying selection. This has resulted in complete minor intron loss in at least nine eukaryotic lineages. However, minor introns are highly conserved in land plants and metazoans, where their importance is underscored by embryonic lethality when the minor spliceosome is inactivated. Conditional inactivation of the minor spliceosome has shown that rapidly dividing progenitor cells are highly sensitive to minor spliceosome loss. Indeed, we found that MIGs were significantly enriched in a screen for genes essential for survival in 341 cycling cell lines. Here, we propose that minor introns inserted randomly into genes in LECA or earlier and were subsequently conserved in genes crucial for cycling cell survival. We hypothesize that the essentiality of MIGs allowed minor introns to endure through the unicellularity of early eukaryotic evolution. Moreover, we identified 59 MIGs that emerged after LECA, and that many of these are essential for cycling cell survival, reinforcing our essentiality model for MIG conservation. This suggests that minor intron emergence is dynamic across eukaryotic evolution, and that minor introns should not be viewed as molecular fossils. We also posit that minor intron splicing was co-opted in multicellular evolution as a regulatory switch for en masse control of MIG expression and the biological processes they regulate. Specifically, this mode of regulation could control cell proliferation and thus body size, an idea supported by domestication syndrome, wherein MIGs are enriched in common candidate animal domestication genes.
Collapse
Affiliation(s)
- Marybeth Baumgartner
- Department of Physiology and Neurobiology, University of Connecticut, Mansfield, CT, United States.,Institute of Brain and Cognitive Sciences, University of Connecticut, Mansfield, CT, United States
| | - Kyle Drake
- Department of Physiology and Neurobiology, University of Connecticut, Mansfield, CT, United States
| | - Rahul N Kanadia
- Department of Physiology and Neurobiology, University of Connecticut, Mansfield, CT, United States.,Institute of Systems Genomics, University of Connecticut, Mansfield, CT, United States
| |
Collapse
|
33
|
Francis L, Greco KV, Boccaccini AR, Roether JJ, English NR, Huang H, Ploeg R, Ansari T. Development of a novel hybrid bioactive hydrogel for future clinical applications. J Biomater Appl 2019; 33:447-465. [PMID: 30223736 DOI: 10.1177/0885328218794163] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Three-dimensional hydrogels are ideal for tissue engineering applications due to their structural integrity and similarity to native soft tissues; however, they can lack mechanical stability. Our objective was to develop a bioactive and mechanically stable hydrogel for clinical application. Auricular cartilage was decellularised using a combination of hypertonic and hypotonic solutions with and without enzymes to produce acellular tissue. Methacryloyl groups were crosslinked with alginate and PVA main chains via 2-aminoethylmathacrylate and the entire macromonomer further crosslinked with the acellular tissue. The resultant hydrogels were characterised for its physicochemical properties (using NMR), in vitro degradation (via GPC analysis), mechanical stability (compression tests) and in vitro biocompatibility (co-culture with bone marrow-derived mesenchymal stem cells). Following decellularisation, the cartilage tissue showed to be acellular at a significant level (DNA content 25.33 ng/mg vs. 351.46 ng/mg control tissue), with good structural and molecular integrity of the retained extra cellular matrix (s-GAG= 0.19 μg/mg vs. 0.65 μg/mg ±0.001 control tissue). Proteomic analysis showed that collagen subtypes and proteoglycans were retained, and SEM and TEM showed preserved matrix ultra-structure. The hybrid hydrogel was successfully cross-linked with biological and polymer components, and it was stable for 30 days in simulated body fluid (poly dispersal index for alginate with tissue was stable at 1.08 and for PVA with tissue was stable at 1.16). It was also mechanically stable (Young's modulus of 0.46 ± 0.31 KPa) and biocompatible, as it was able to support the development of a multi-cellular feature with active cellular proliferation in vitro. We have shown that it is possible to successfully combine biological tissue with both a synthetic and natural polymer and create a hybrid bioactive hydrogel for clinical application.
Collapse
Affiliation(s)
- Lydia Francis
- 1 Tissue Engineering and Regenerative Medicine, Northwick Park Institute for Medical Research (NPIMR), Harrow, UK
| | - Karin V Greco
- 1 Tissue Engineering and Regenerative Medicine, Northwick Park Institute for Medical Research (NPIMR), Harrow, UK
| | - Aldo R Boccaccini
- 2 Department of Materials Science, Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Judith J Roether
- 2 Department of Materials Science, Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Nicholas R English
- 3 Antigen Presentation Research Group, Imperial College London, London, UK
| | - Honglei Huang
- 4 Nuffield Department of Surgical Sciences, Oxford Transplant Centre, University of Oxford, Oxford, UK
| | - R Ploeg
- 4 Nuffield Department of Surgical Sciences, Oxford Transplant Centre, University of Oxford, Oxford, UK
| | - Tahera Ansari
- 1 Tissue Engineering and Regenerative Medicine, Northwick Park Institute for Medical Research (NPIMR), Harrow, UK
| |
Collapse
|
34
|
Zacchigna S, Martinelli V, Moimas S, Colliva A, Anzini M, Nordio A, Costa A, Rehman M, Vodret S, Pierro C, Colussi G, Zentilin L, Gutierrez MI, Dirkx E, Long C, Sinagra G, Klatzmann D, Giacca M. Paracrine effect of regulatory T cells promotes cardiomyocyte proliferation during pregnancy and after myocardial infarction. Nat Commun 2018; 9:2432. [PMID: 29946151 PMCID: PMC6018668 DOI: 10.1038/s41467-018-04908-z] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 05/16/2018] [Indexed: 02/07/2023] Open
Abstract
Cardiomyocyte proliferation stops at birth when the heart is no longer exposed to maternal blood and, likewise, to regulatory T cells (Tregs) that are expanded to promote maternal tolerance towards the fetus. Here, we report a role of Tregs in promoting cardiomyocyte proliferation. Treg-conditioned medium promotes cardiomyocyte proliferation, similar to the serum from pregnant animals. Proliferative cardiomyocytes are detected in the heart of pregnant mothers, and Treg depletion during pregnancy decreases both maternal and fetal cardiomyocyte proliferation. Treg depletion after myocardial infarction results in depressed cardiac function, massive inflammation, and scarce collagen deposition. In contrast, Treg injection reduces infarct size, preserves contractility, and increases the number of proliferating cardiomyocytes. The overexpression of six factors secreted by Tregs (Cst7, Tnfsf11, Il33, Fgl2, Matn2, and Igf2) reproduces the therapeutic effect. In conclusion, Tregs promote fetal and maternal cardiomyocyte proliferation in a paracrine manner and improve the outcome of myocardial infarction. Regulatory T cells (Tregs) expand during pregnancy to promote tolerance towards the fetus. Here the authors show that Tregs induce proliferation of fetal and maternal cardiomyocytes during pregnancy and enhance myocardial repair via proliferation-promoting paracrine actions.
Collapse
Affiliation(s)
- Serena Zacchigna
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149, Trieste, Italy. .,Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy and Center for Translational Cardiology, Azienda Sanitaria Universitaria Integrata di Trieste, 34129, Trieste, Italy.
| | - Valentina Martinelli
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149, Trieste, Italy
| | - Silvia Moimas
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy and Center for Translational Cardiology, Azienda Sanitaria Universitaria Integrata di Trieste, 34129, Trieste, Italy.,Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149, Trieste, Italy
| | - Andrea Colliva
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149, Trieste, Italy
| | - Marco Anzini
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy and Center for Translational Cardiology, Azienda Sanitaria Universitaria Integrata di Trieste, 34129, Trieste, Italy
| | - Andrea Nordio
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy and Center for Translational Cardiology, Azienda Sanitaria Universitaria Integrata di Trieste, 34129, Trieste, Italy
| | - Alessia Costa
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149, Trieste, Italy
| | - Michael Rehman
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149, Trieste, Italy
| | - Simone Vodret
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149, Trieste, Italy
| | - Cristina Pierro
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149, Trieste, Italy
| | - Giulia Colussi
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149, Trieste, Italy
| | - Lorena Zentilin
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149, Trieste, Italy
| | - Maria Ines Gutierrez
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149, Trieste, Italy
| | - Ellen Dirkx
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149, Trieste, Italy
| | - Carlin Long
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149, Trieste, Italy
| | - Gianfranco Sinagra
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy and Center for Translational Cardiology, Azienda Sanitaria Universitaria Integrata di Trieste, 34129, Trieste, Italy
| | - David Klatzmann
- Sorbonne Université, UPMC Univ Paris 06, INSERM, UMR_S 959, Immunology-Immunopathology-Immunotherapy (i3), F-75005, Paris, France.,AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Department of Biotherapies, Clinical Investigation Center in Biotherapy and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), F-75013, Paris, France
| | - Mauro Giacca
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy and Center for Translational Cardiology, Azienda Sanitaria Universitaria Integrata di Trieste, 34129, Trieste, Italy.,Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149, Trieste, Italy
| |
Collapse
|
35
|
Cheng A, Cain SA, Tian P, Baldwin AK, Uppanan P, Kielty CM, Kimber SJ. Recombinant Extracellular Matrix Protein Fragments Support Human Embryonic Stem Cell Chondrogenesis. Tissue Eng Part A 2018; 24:968-978. [PMID: 29279011 PMCID: PMC5984563 DOI: 10.1089/ten.tea.2017.0285] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
We previously developed a 14-day culture protocol under potentially GMP, chemically defined conditions, to generate chondroprogenitors from human embryonic stem cells (hESCs). In vivo work has confirmed the cartilage repair capacity of these cells in a nude rat osteochondral defect model. Aiming to enhance hESC-chondrogenesis, we screened a range of extracellular matrix (ECM) molecules for their ability to support differentiation of hESCs toward chondrocytes. We identified two novel ECM protein fragments that supported hESC-chondrogenesis: Fibronectin III (fibronectin 7-14 protein fragments, including the RGD domain, syndecan-binding domain, and heparin-binding domain) and fibrillin-1 (FBN1) fragment PF8 (encoded by exons 30-38, residues 1238-1605, which contains the RGD motif but not heparin-binding site). These two protein fragments support hESC-chondrogenesis compared with the substrates routinely used previously (a mixture of fibronectin and gelatin) in our directed chondrogenic protocol. We have identified recombinant fibronectin fragment (FN III) and FBNI fragment (PF8) as alternative coating substrates to promote expression of genes known to regulate chondrocytes and code for chondrocyte ECM components. These recombinant protein fragments are likely to have better batch to batch stability than full-length molecules, especially where extracted from tissue/serum.
Collapse
Affiliation(s)
- Aixin Cheng
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Stuart A. Cain
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Pinyuan Tian
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Andrew K. Baldwin
- Academic Group—Engineering, Sports and Sciences, The University of Bolton, Bolton, United Kingdom
| | | | - Cay M. Kielty
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Susan J. Kimber
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
36
|
Hawkins AG, Basrur V, da Veiga Leprevost F, Pedersen E, Sperring C, Nesvizhskii AI, Lawlor ER. The Ewing Sarcoma Secretome and Its Response to Activation of Wnt/beta-catenin Signaling. Mol Cell Proteomics 2018; 17:901-912. [PMID: 29386236 PMCID: PMC5930412 DOI: 10.1074/mcp.ra118.000596] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Indexed: 12/11/2022] Open
Abstract
Tumor: tumor microenvironment (TME) interactions are critical for tumor progression and the composition and structure of the local extracellular matrix (ECM) are key determinants of tumor metastasis. We recently reported that activation of Wnt/beta-catenin signaling in Ewing sarcoma cells induces widespread transcriptional changes that are associated with acquisition of a metastatic tumor phenotype. Significantly, ECM protein-encoding genes were found to be enriched among Wnt/beta-catenin induced transcripts, leading us to hypothesize that activation of canonical Wnt signaling might induce changes in the Ewing sarcoma secretome. To address this hypothesis, conditioned media from Ewing sarcoma cell lines cultured in the presence or absence of Wnt3a was collected for proteomic analysis. Label-free mass spectrometry was used to identify and quantify differentially secreted proteins. We then used in silico databases to identify only proteins annotated as secreted. Comparison of the secretomes of two Ewing sarcoma cell lines revealed numerous shared proteins, as well as a degree of heterogeneity, in both basal and Wnt-stimulated conditions. Gene set enrichment analysis of secreted proteins revealed that Wnt stimulation reproducibly resulted in increased secretion of proteins involved in ECM organization, ECM receptor interactions, and collagen formation. In particular, Wnt-stimulated Ewing sarcoma cells up-regulated secretion of structural collagens, as well as matricellular proteins, such as the metastasis-associated protein, tenascin C (TNC). Interrogation of published databases confirmed reproducible correlations between Wnt/beta-catenin activation and TNC and COL1A1 expression in patient tumors. In summary, this first study of the Ewing sarcoma secretome reveals that Wnt/beta-catenin activated tumor cells upregulate secretion of ECM proteins. Such Wnt/beta-catenin mediated changes are likely to impact on tumor: TME interactions that contribute to metastatic progression.
Collapse
Affiliation(s)
| | | | | | | | | | - Alexey I Nesvizhskii
- §Pathology, and
- ¶Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | | |
Collapse
|
37
|
Doxorubicin-triggered self-assembly of native amphiphilic peptides into spherical nanoparticles. Oncotarget 2018; 7:58445-58458. [PMID: 27533248 PMCID: PMC5295442 DOI: 10.18632/oncotarget.11213] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 07/19/2016] [Indexed: 01/30/2023] Open
Abstract
In this study, we designed and fabricated self-assembly nanospheres, which consisted of a P45 peptide and doxorubicin (Dox). P45 is a hybrid peptide composed of an Arg-Gly-Asp motif linked to the human matrilin-1 C-terminal domain by a serine linker. The fabricated nanospheres had a uniform mulberry-like spherical shape, a diameter of 63 nm, excellent polydispersity, and high Dox drug-loading efficiency. In the presence of the RGD motif, the Dox/P45 nanospheres could specifically target A549 cells, which have high integrin αvβ3 expression. Confocal laser scanning microscopy and flow cytometry results showed the enhanced cellular uptake of Dox/P45, and the CCK8 assay indicated the low cytotoxicity of the nanospheres to normal human embryonic kidney 293 cells. Furthermore, the fabricated nanospheres were stable in a physiological environment, but they disassembled and exhibited a rapid Dox release in an acidic atmosphere, allowing for a specific pH-sensitive release into cytosol after cellular uptake. These results suggest that natural amphiphilic peptides can be used as carriers of nanodrugs for targeting delivery as well as controlled drug release for cancer therapy.
Collapse
|
38
|
Wu PL, He YF, Yao HH, Hu B. Martrilin-3 (MATN3) Overexpression in Gastric Adenocarcinoma and its Prognostic Significance. Med Sci Monit 2018; 24:348-355. [PMID: 29343680 PMCID: PMC5784332 DOI: 10.12659/msm.908447] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND The aim of this study was to investigate the expression level of martrilin-3 (MATN3) in patients with gastric adenocarcinoma (GAC) and to investigate the prognostic significance of MATN3. MATERIAL AND METHODS Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) data were used to predict the expression and prognostic value of MATN3 mRNA in GAC patients. Seventy-six GAC patients had GAC tissue samples and paired adjacent normal tissue samples collected retrospectively to examine the MATN3 protein expression level by immunohistochemical staining. Furthermore, Kaplan-Meier univariate and Cox multivariate analyses were used to verify the correlation between MATN3 expression and clinicopathological parameters of GAC patients and the prognostic significance of MATN3. RESULTS The GEO and TCGA data predicted that MATN3 mRNA levels were significantly higher in GAC tissue compared to normal tissue (all p<0.05). Further survival analyses showed that GAC patients with high mRNA expression of MATN3 had significantly lower disease-free survival (DFS) and overall survival (OS) time than those with low mRNA expression of MATN3 (all p<0.05). Subsequent immunohistochemical staining results confirmed that the MATN3 protein levels in GAC tissues were highly expressed (p=0.000) compared to normal tissues. In addition, GAC patients with high protein expression of MATN3 had remarkably decreased OS compared to patients with low protein expression of MATN3 (p=0.000). Univariate and multivariate survival analyses revealed that MATN3 high expression could be used as an independent predictor of poor prognosis in GAC patients (all p=0.000). CONCLUSIONS This study confirmed that MATN3 protein was highly expressed in GAC patients, and MATN3 overexpression could be used as an independent predictor of poor prognosis in GAC patients.
Collapse
Affiliation(s)
- Ping-Li Wu
- Department of Medical Oncology, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui, China (mainland).,Department of Medical Oncology, Suixi County Hospital, Huaibei, Anhui, China (mainland)
| | - Yi-Fu He
- Department of Medical Oncology, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui, China (mainland)
| | - Han-Hui Yao
- Department of General Surgery, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui, China (mainland)
| | - Bing Hu
- Department of Medical Oncology, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui, China (mainland)
| |
Collapse
|
39
|
Junkunlo K, Söderhäll K, Söderhäll I. Clotting protein - An extracellular matrix (ECM) protein involved in crustacean hematopoiesis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 78:132-140. [PMID: 28943319 DOI: 10.1016/j.dci.2017.09.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/19/2017] [Accepted: 09/19/2017] [Indexed: 06/07/2023]
Abstract
Hematopoietic progenitor cells in crustaceans are organized in lobule-like structures surrounded by different types of cells and extracellular matrix (ECM) proteins in a Hematopoietic tissue (HPT). Here we show that the clotting protein (CP) is part of the ECM in HPT and is secreted during HPT cell culture. The formation of a filamentous network of CP was observed in HPT cell culture. A high amount of CP protein was detected at the surfaces of undifferentiated cells (round-shaped) compared with migrating cells (spindle shaped). Co-localization of the CP protein and TGase activity was observed on the cell surface and filamentous network between cells. A role for CP together with collagen was revealed in a 3D culture in which a collagen-I matrix was immobilized with CP or supplemented with CP. The results showed possible functions of CP, collagen, TGase and the cytokine Ast1 in the regulation of HPT progenitor cell behavior. This is the first study to provide insight into the role of CP, which probably not only participates in clot formation but also functions as an ECM component protein controlling hematopoietic stem cell behavior.
Collapse
Affiliation(s)
- Kingkamon Junkunlo
- Department of Comparative Physiology, Uppsala University, Norbyvägen 18A, 752 36 Uppsala, Sweden
| | - Kenneth Söderhäll
- Department of Comparative Physiology, Uppsala University, Norbyvägen 18A, 752 36 Uppsala, Sweden
| | - Irene Söderhäll
- Department of Comparative Physiology, Uppsala University, Norbyvägen 18A, 752 36 Uppsala, Sweden.
| |
Collapse
|
40
|
Abstract
Marilins mediate interactions between macromolecular components of the extracellular matrix, e.g., collagens and proteoglycans. They are composed of von Willebrand factor type A and epidermal growth factor-like domains and the subunits oligomerize via coiled-coil domains. Matrilin-1 and -3 are abundant in hyaline cartilage, whereas matrilin-2 and -4 are widespread but less abundant. Mutations in matrilin genes have been linked to chondrodysplasias and osteoarthritis and recently characterization of matrilin-deficient mice revealed novel functions in mechanotransduction, regeneration, or inflammation. Due to their intrinsic adhesiveness and partially also low abundance, the study of matrilins is cumbersome. In this chapter, we describe methods for purification of matrilins from tissue, analysis of matrilins in tissue extracts, recombinant expression, and generation of matrilin-specific antibodies.
Collapse
Affiliation(s)
- Mats Paulsson
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Cologne Center for Musculoskeletal Biomechanics (CCMB), University of Cologne, Cologne, Germany
| | - Raimund Wagener
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany; Cologne Center for Musculoskeletal Biomechanics (CCMB), University of Cologne, Cologne, Germany.
| |
Collapse
|
41
|
Muttigi MS, Kim BJ, Choi B, Yoshie A, Kumar H, Han I, Park H, Lee SH. Matrilin-3 codelivery with adipose-derived mesenchymal stem cells promotes articular cartilage regeneration in a rat osteochondral defect model. J Tissue Eng Regen Med 2017; 12:667-675. [PMID: 28556569 DOI: 10.1002/term.2485] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 05/12/2017] [Accepted: 05/19/2017] [Indexed: 12/14/2022]
Abstract
Matrilin-3 is an essential extracellular matrix component present only in cartilaginous tissues. Matrilin-3 exerts chondroprotective effects by regulating an anti-inflammatory function and extracellular matrix components. We hypothesized that the codelivery of matrilin-3 with infrapatellar adipose-tissue-derived mesenchymal stem cells (Ad-MSCs) may enhance articular cartilage regeneration. Matrilin-3 treatment of Ad-MSCs in serum-free media induced collagen II and aggrecan expression, and matrilin-3 in chondrogenic media also enhanced in vitro chondrogenic differentiation. Next, the in vivo effect of matrilin-3 codelivery with Ad-MSCs on cartilage regeneration was assessed in an osteochondral defect model in Sprague Dawley rats: Ad-MSCs and hyaluronic acid were implanted at the defect site with or without matrilin-3 (140, 280, and 700 ng). Safranin O staining revealed that matrilin-3 (140 and 280 ng) treatment significantly improved cartilage regeneration and glycosaminoglycan accumulation. In the animals treated with 140-ng matrilin-3, in particular, the defect site exhibited complete integration with surrounding tissue and a smooth glistening surface. The International Cartilage Repair Society macroscopic and O'Driscoll microscopic scores for regenerated cartilage were furthermore shown to be considerably higher for this group (matrilin-3; 140 ng) compared with the other groups. Furthermore, the defects treated with 140-ng matrilin-3 revealed significant hyaline-like cartilage regeneration in the osteochondral defect model; in contrast, the defects treated with 700-ng matrilin-3 exhibited drastically reduced cartilage regeneration with mixed hyaline-fibrocartilage morphology. Codelivery of matrilin-3 with Ad-MSCs significantly influenced articular cartilage regeneration, supporting the potential use of this tissue-specific protein for a cartilage-targeted stem cell therapy.
Collapse
Affiliation(s)
- Manjunatha S Muttigi
- Department of Biomedical Science, CHA University, Seongnam, South Korea.,School of Integrative Engineering, Chung-Ang University, Seoul, South Korea
| | - Byoung Ju Kim
- Department of Biomedical Science, CHA University, Seongnam, South Korea
| | - Bogyu Choi
- Department of Biomedical Science, CHA University, Seongnam, South Korea
| | - Arai Yoshie
- Department of Biomedical Science, CHA University, Seongnam, South Korea
| | - Hemant Kumar
- Department of Biomedical Science, CHA University, Seongnam, South Korea.,Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Inbo Han
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul, South Korea
| | - Soo-Hong Lee
- Department of Biomedical Science, CHA University, Seongnam, South Korea
| |
Collapse
|
42
|
Smith AM, Papaleo C, Reid CW, Bliss JM. RNA-Seq reveals a central role for lectin, C1q and von Willebrand factor A domains in the defensive glue of a terrestrial slug. BIOFOULING 2017; 33:741-754. [PMID: 28899232 PMCID: PMC6124484 DOI: 10.1080/08927014.2017.1361413] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 07/25/2017] [Indexed: 06/07/2023]
Abstract
The tough, hydrogel glue produced by the slug Arion subfuscus achieves impressive performance through metal-based, protein cross-links. The primary sequence of these proteins was determined through transcriptome sequencing and proteome analysis by tandem mass spectrometry. The main proteins that correlate with adhesive function are a group of 11 small, highly abundant lectin-like proteins. These proteins matched the ligand-binding C-lectin, C1q or H-lectin domains. The variety of different lectin-like proteins and their potential for oligomerization suggests that they function as versatile and potent cross-linkers. In addition, the glue contains five matrilin-like proteins that are rich in von Willebrand factor A (VWA) and EGF domains. Both C-lectins and VWA domains are known to bind to ligands using divalent cations. These findings are consistent with the double network mechanism proposed for slug glue, with divalent ions serving as sacrificial bonds to dissipate energy.
Collapse
Affiliation(s)
- Andrew M. Smith
- Ithaca College, Department of Biology, 953 Danby Road, Ithaca, NY 14850 607-274-3975, ,
| | - Cassandra Papaleo
- Ithaca College, Department of Biology, 953 Danby Road, Ithaca, NY 14850 607-274-3975, ,
| | - Christopher W. Reid
- Bryant University, Science and Technology Department, 1150 Douglas Pike, Smithfield, RI 02917 401-232-6000,
| | - Joseph M. Bliss
- Women & Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University 100 Dudley Street, Providence, RI 02905, 401-274-1100,
| |
Collapse
|
43
|
Diab SM, Kamal HM, Mansour AI, Fawzy RM, Azab BS. Clinical significance of Matrilin-3 gene polymorphism in Egyptian patients with primary knee osteoarthritis. Eur J Rheumatol 2017; 4:200-204. [PMID: 28983412 PMCID: PMC5621842 DOI: 10.5152/eurjrheum.2016.16107] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 03/05/2017] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE Osteoarthritis (OA) is a multifactorial, degenerative, and inflammatory disorder of joints causing damage of the articular cartilage, formation of osteophytes, and eburination of the subchondral bone. Matrilin-3 (MATN-3) is a non-collagenous oligomeric extracellular matrix protein (ECM), which is the smallest member of the matrilin family. This study was conducted to identify the potential association and clinical significance of MATN-3 rs8176070 (SNP6) polymorphism in a series of Egyptian patients with primary knee OA. MATERIAL AND METHODS Polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) was used to determine genotypes of MATN-3 SNP6 for 50 primary knee OA patients in addition to 50 healthy subjects of the same sex and age range. Full history was obtained from OA patients, followed by clinical examination, together with clinical assessment of the severity of knee OA using Lequesne Algofunctional Index score and radiological grading using the Kellgren-Lawrence grade scale (KL). RESULTS With regard to genotypes of MATN-3 gene SNP6 (rs8176070), a statistically significant difference between OA patients and healthy control subjects was found for the B\b genotype and b allele (p=0.046 and 0.042 respectively), with the prevalence being higher in OA patients with a high risk to develop OA (Odds Ratio [OR]=2.250, 95% CI=1.011-5.008). Patients with the B\b genotype had worse clinical and radiological findings than those with B\B and b\b genotypes. CONCLUSION The investigated polymorphism in the MATN-3 gene may reflect the risk and severity of knee OA in Egyptian patients, particularly with the B\b genotype.
Collapse
Affiliation(s)
- Safia M. Diab
- Department of Clinical & Chemical Pathology, Benha University School Medicine, Benha, Egypt
| | - Howyda M. Kamal
- Department of Clinical & Chemical Pathology, Benha University School Medicine, Benha, Egypt
| | - Amira I. Mansour
- Department of Clinical & Chemical Pathology, Benha University School Medicine, Benha, Egypt
| | - Rasha M. Fawzy
- Department of Rheumatology & Rehabilitation, Benha University School Medicine, Benha, Egypt
| | - Basma S. Azab
- Department of Clinical & Chemical Pathology, Benha University School Medicine, Benha, Egypt
| |
Collapse
|
44
|
Firner S, Zaucke F, Michael J, Dargel J, Schiwy-Bochat KH, Heilig J, Rothschild MA, Eysel P, Brüggemann GP, Niehoff A. Extracellular Distribution of Collagen II and Perifibrillar Adapter Proteins in Healthy and Osteoarthritic Human Knee Joint Cartilage. J Histochem Cytochem 2017; 65:593-606. [PMID: 28846474 DOI: 10.1369/0022155417729154] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Perifibrillar adapter proteins, interconnecting collagen fibrils, and linking the collagen network with the aggrecan matrix seem to play a crucial role in the pathogenesis of osteoarthritis (OA). Therefore, we examined immunohistochemically the extracellular distribution of collagen II and the main perifibrillar adapter proteins-collagen IX, decorin, cartilage oligomeric matrix protein (COMP), and matrilin-3-in human samples of healthy (n=4) and OA (n=42) knee joint cartilage. Histopathology assessment was performed using an OA score. Staining patterns were evaluated in relation to the disease stage. The perifibrillar adapter proteins were uniformly distributed in the upper zones of healthy cartilage. In moderate OA (n=8; score 14.3 ± 4.7), all proteins analyzed were locally absent in the fibrillated area or the superficial and upper mid zone. In advanced OA (n=20; score 18.9 ± 5.3), they were uniformly distributed in these zones and accumulated pericellularly. Perifibrillar adapter proteins are important for the stabilization of the collagen network in the upper zones of healthy cartilage. Their degradation might be a critical event in early OA. In advanced OA, there are indications for an increased synthesis in an attempt to regenerate the lost tissue and to protect the remaining cartilage from further destruction.
Collapse
Affiliation(s)
- Sara Firner
- Institute of Biomechanics and Orthopaedics, German Sport University Cologne, Cologne, Germany
| | - Frank Zaucke
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopaedic University Hospital Friedrichsheim gGmbH, Frankfurt, Germany.,Cologne Center for Musculoskeletal Biomechanics (CCMB), Medical Faculty, University of Cologne, Cologne, Germany
| | - Joern Michael
- Department of Orthopaedic and Trauma Surgery, University Hospital Cologne, Cologne, Germany
| | - Jens Dargel
- Department of Orthopaedic and Trauma Surgery, University Hospital Cologne, Cologne, Germany
| | | | - Juliane Heilig
- Cologne Center for Musculoskeletal Biomechanics (CCMB), Medical Faculty, University of Cologne, Cologne, Germany
| | | | - Peer Eysel
- Department of Orthopaedic and Trauma Surgery, University Hospital Cologne, Cologne, Germany.,Cologne Center for Musculoskeletal Biomechanics (CCMB), Medical Faculty, University of Cologne, Cologne, Germany
| | - Gert-Peter Brüggemann
- Institute of Biomechanics and Orthopaedics, German Sport University Cologne, Cologne, Germany.,Cologne Center for Musculoskeletal Biomechanics (CCMB), Medical Faculty, University of Cologne, Cologne, Germany
| | - Anja Niehoff
- Institute of Biomechanics and Orthopaedics, German Sport University Cologne, Cologne, Germany.,Cologne Center for Musculoskeletal Biomechanics (CCMB), Medical Faculty, University of Cologne, Cologne, Germany
| |
Collapse
|
45
|
Woltersdorf C, Bonk M, Leitinger B, Huhtala M, Käpylä J, Heino J, Gil Girol C, Niland S, Eble JA, Bruckner P, Dreier R, Hansen U. The binding capacity of α1β1-, α2β1- and α10β1-integrins depends on non-collagenous surface macromolecules rather than the collagens in cartilage fibrils. Matrix Biol 2017; 63:91-105. [PMID: 28192200 DOI: 10.1016/j.matbio.2017.02.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 02/03/2017] [Accepted: 02/04/2017] [Indexed: 12/30/2022]
Abstract
Interactions of cells with supramolecular aggregates of the extracellular matrix (ECM) are mediated, in part, by cell surface receptors of the integrin family. These are important molecular components of cell surface-suprastructures regulating cellular activities in general. A subfamily of β1-integrins with von Willebrand-factor A-like domains (I-domains) in their α-chains can bind to collagen molecules and, therefore, are considered as important cellular mechano-receptors. Here we show that chondrocytes strongly bind to cartilage collagens in the form of individual triple helical molecules but very weakly to fibrils formed by the same molecules. We also find that chondrocyte integrins α1β1-, α2β1- and α10β1-integrins and their I-domains have the same characteristics. Nevertheless we find integrin binding to mechanically generated cartilage fibril fragments, which also comprise peripheral non-collagenous material. We conclude that cell adhesion results from binding of integrin-containing adhesion suprastructures to the non-collagenous fibril periphery but not to the collagenous fibril cores. The biological importance of the well-investigated recognition of collagen molecules by integrins is unknown. Possible scenarios may include fibrillogenesis, fibril degradation and/or phagocytosis, recruitment of cells to remodeling sites, or molecular signaling across cytoplasmic membranes. In these circumstances, collagen molecules may lack a fibrillar organization. However, other processes requiring robust biomechanical functions, such as fibril organization in tissues, cell division, adhesion, or migration, do not involve direct integrin-collagen interactions.
Collapse
Affiliation(s)
- Christian Woltersdorf
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, D-48149 Münster, Germany
| | - Melanie Bonk
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, D-48149 Münster, Germany
| | - Birgit Leitinger
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Mikko Huhtala
- University of Turku, Department of Biochemistry, FI-20014 Turun Yliopisto, Finland
| | - Jarmo Käpylä
- University of Turku, Department of Biochemistry, FI-20014 Turun Yliopisto, Finland
| | - Jyrki Heino
- University of Turku, Department of Biochemistry, FI-20014 Turun Yliopisto, Finland
| | - Christian Gil Girol
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, D-48149 Münster, Germany
| | - Stephan Niland
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, D-48149 Münster, Germany
| | - Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, D-48149 Münster, Germany
| | - Peter Bruckner
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, D-48149 Münster, Germany.
| | - Rita Dreier
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, D-48149 Münster, Germany
| | - Uwe Hansen
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, D-48149 Münster, Germany; Institute of Experimental Musculoskeletal Medicine (IEMM), University of Münster, DE-48149 Münster, Germany
| |
Collapse
|
46
|
Abstract
The zebrafish skeleton shares many similarities with human and other vertebrate skeletons. Over the past years, work in zebrafish has provided an extensive understanding of the basic developmental mechanisms and cellular pathways directing skeletal development and homeostasis. This review will focus on the cell biology of cartilage and bone and how the basic cellular processes within chondrocytes and osteocytes function to assemble the structural frame of a vertebrate body. We will discuss fundamental functions of skeletal cells in production and secretion of extracellular matrix and cellular activities leading to differentiation of progenitors to mature cells that make up the skeleton. We highlight important examples where findings in zebrafish provided direction for the search for genes causing human skeletal defects and also how zebrafish research has proven important for validating candidate human disease genes. The work we cover here illustrates utility of zebrafish in unraveling molecular mechanisms of cellular functions necessary to form and maintain a healthy skeleton.
Collapse
Affiliation(s)
- Lauryn N Luderman
- Vanderbilt University Medical Center, Nashville, TN, United States; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, United States; Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, United States
| | - Gokhan Unlu
- Vanderbilt University Medical Center, Nashville, TN, United States; Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, United States; Vanderbilt University, Nashville, TN, United States
| | - Ela W Knapik
- Vanderbilt University Medical Center, Nashville, TN, United States; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, United States; Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, United States; Vanderbilt University, Nashville, TN, United States.
| |
Collapse
|
47
|
Sato N, Taniguchi T, Goda Y, Kosaka H, Higashino K, Sakai T, Katoh S, Yasui N, Sairyo K, Taniguchi H. Proteomic Analysis of Human Tendon and Ligament: Solubilization and Analysis of Insoluble Extracellular Matrix in Connective Tissues. J Proteome Res 2016; 15:4709-4721. [DOI: 10.1021/acs.jproteome.6b00806] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Nori Sato
- Department
of Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Takako Taniguchi
- Division
of Disease Proteomics, Institute for Enzyme Research, Tokushima University, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yuichiro Goda
- Department
of Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Hirofumi Kosaka
- Department
of Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Kosaku Higashino
- Department
of Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Toshinori Sakai
- Department
of Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Shinsuke Katoh
- Department
of Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Natsuo Yasui
- Department
of Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Koichi Sairyo
- Department
of Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Hisaaki Taniguchi
- Division
of Disease Proteomics, Institute for Enzyme Research, Tokushima University, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| |
Collapse
|
48
|
Yilmaz AD, Yazicioğlu D, Tuzuner Oncul MA, Ereş G, Sayan NB. Association of Matrilin-3 Gene Polymorphism with Temporomandibular Joint Internal Derangement. Genet Test Mol Biomarkers 2016; 20:563-568. [PMID: 27533128 DOI: 10.1089/gtmb.2016.0037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Temporomandibular joint internal derangement (TMJ ID) is a multifactorial complex disease characterised by articular disc degeneration. Matrilin-3 is a cartilage and bone-specific adaptor protein, and amino-acid substitutions in the protein are associated with skeletal diseases and joint disorders. We aimed to detect the variants of Matrilin-3 gene (MATN3) in a TMJ ID case-control group and to investigate the risk association of the detected variants with TMJ ID. MATERIALS AND METHODS A case control study was conducted consisting of 57 unrelated TMJ ID patients (32.7 ± 8.2) and 96 unrelated healthy controls (26.63 ± 3.05) without TMJ ID to look for associations with variants of the MATN3 gene. DNA from individual subjects was extracted and each of the eight exons was amplified by polymerase chain reaction using and analyzed by single-strand conformational polymorphism (SSCP) analysis. SSCP variants were subjected to DNA sequence analysis, which yielded band pattern variations in exon 2 of the gene. We further analyzed exon 2 by DNA sequencing to determine the sequence of these variants. RESULTS We identified SSCP band patterns variants in exon 2 of the MATN3 gene which upon sequencing revealed a single C to T transition mutation (rs28598872) c.447 C>T (g.11608 C>T). This polymorphism is predicted to result in a synonymous mutation (pAla149 = ). The TT and CT genotypes were more prevalent than the CC genotype in TMJ ID patients compared to the control group with a risk factor of 2.12 (confidence intervals [CI] :0.88-5.08) and 2.0 (CI:0.726-5.508). In addition, TMJ ID patients were divided into two groups as anterior disc displacement with reduction (ADDWR) and anterior disc displacement without reduction (ADDWOR) and compared with the controls. The TT and CT genotypes were more prevalent than the CC genotype in ADDWR patients compared to the control group with a risk factor of 3.85 (CI:0.927-16.048) and 3.75 (1.02-13.786), respectively. We found that, among ADDWR patients, the T allele is a risk factor both in homozygous and heterozygous carriers (p < 0.052, p < 0.036). CONCLUSION The results of the study indicate a potential role for the MATN3 rs28598872 polymorphism in the pathogenesis of TMJ ID.
Collapse
Affiliation(s)
- Ayça Dilara Yilmaz
- 1 Molecular Biology Laboratory, Faculty of Dentistry, Ankara University , Ankara, Turkey
| | - Duygu Yazicioğlu
- 2 Private Practice , Oral and Maxillofacial Surgery, Ankara, Turkey
| | - Mine Aysegul Tuzuner Oncul
- 3 Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Ankara University , Ankara, Turkey
| | - Gülden Ereş
- 4 Department of Periodontology, Faculty of Dentistry, Ankara University , Ankara, Turkey
| | - Nejat Bora Sayan
- 2 Private Practice , Oral and Maxillofacial Surgery, Ankara, Turkey
| |
Collapse
|
49
|
Uckelmann H, Blaszkiewicz S, Nicolae C, Haas S, Schnell A, Wurzer S, Wagener R, Aszodi A, Essers MAG. Extracellular matrix protein Matrilin-4 regulates stress-induced HSC proliferation via CXCR4. J Exp Med 2016; 213:1961-71. [PMID: 27573814 PMCID: PMC5030798 DOI: 10.1084/jem.20151713] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 07/07/2016] [Indexed: 01/20/2023] Open
Abstract
Essers et al. find that the extracellular matrix adaptor protein Matrilin-4 confers a resistance to stress stimuli in hematopoietic stem cells. During homeostasis, hematopoietic stem cells (HSCs) are mostly kept in quiescence with only minor contribution to steady-state hematopoiesis. However, in stress situations such as infection, chemotherapy, or transplantation, HSCs are forced to proliferate and rapidly regenerate compromised hematopoietic cells. Little is known about the processes regulating this stress-induced proliferation and expansion of HSCs and progenitors. In this study, we identified the extracellular matrix (ECM) adaptor protein Matrilin-4 (Matn4) as an important negative regulator of the HSC stress response. Matn4 is highly expressed in long-term HSCs; however, it is not required for HSC maintenance under homeostasis. In contrast, Matn4 is strongly down-regulated in HSCs in response to proliferative stress, and Matn4 deficiency results in increased proliferation and expansion of HSCs and progenitors after myelosuppressive chemotherapy, inflammatory stress, and transplantation. This enhanced proliferation is mediated by a transient down-regulation of CXCR4 in Matn4−/− HSCs upon stress, allowing for a more efficient expansion of HSCs. Thus, we have uncovered a novel link between the ECM protein Matn4 and cytokine receptor CXCR4 involved in the regulation of HSC proliferation and expansion under acute stress.
Collapse
Affiliation(s)
- Hannah Uckelmann
- HSCs and Stress Group, Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69121 Heidelberg, Germany Heidelberg Institute for Stem Cell Technologies and Experimental Medicine gGmbH, 69120 Heidelberg, Germany
| | - Sandra Blaszkiewicz
- HSCs and Stress Group, Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69121 Heidelberg, Germany Heidelberg Institute for Stem Cell Technologies and Experimental Medicine gGmbH, 69120 Heidelberg, Germany
| | - Claudia Nicolae
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Planegg, Germany
| | - Simon Haas
- HSCs and Stress Group, Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69121 Heidelberg, Germany Heidelberg Institute for Stem Cell Technologies and Experimental Medicine gGmbH, 69120 Heidelberg, Germany
| | - Alexandra Schnell
- HSCs and Stress Group, Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69121 Heidelberg, Germany Heidelberg Institute for Stem Cell Technologies and Experimental Medicine gGmbH, 69120 Heidelberg, Germany
| | - Stephan Wurzer
- HSCs and Stress Group, Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69121 Heidelberg, Germany Heidelberg Institute for Stem Cell Technologies and Experimental Medicine gGmbH, 69120 Heidelberg, Germany
| | - Raimund Wagener
- Center for Biochemistry, Medical Faculty and Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Attila Aszodi
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Planegg, Germany Department of General, Trauma, Hand, and Plastic Surgery, Ludwig-Maximilians University, 80539 Munich, Germany
| | - Marieke Alida Gertruda Essers
- HSCs and Stress Group, Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69121 Heidelberg, Germany Heidelberg Institute for Stem Cell Technologies and Experimental Medicine gGmbH, 69120 Heidelberg, Germany
| |
Collapse
|
50
|
Zhang S, Zhang M, Huang H, Zhou S, Du Y, Yi X, Luo J. High glucose-induced Matrilin-2 expression in mouse mesangial cells was mediated by transforming growth factor beta 1 (TGF-β1). Biochem Biophys Res Commun 2016; 474:303-308. [PMID: 27105914 DOI: 10.1016/j.bbrc.2016.04.091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 04/18/2016] [Indexed: 11/28/2022]
Abstract
This study aimed at evaluating the effect of high glucose on the expression of extracellular matrix (ECM) protein Matrilin-2 and the mechanism underlying this effect by using a mouse mesangial cell line. Mouse mesangial cells (MMCs) were cultured in media containing normal (5 mM d-glucose) or high concentrations of glucose (30 mM d-glucose). The expression of Matrilin-2 was assessed by either RT-PCR or western blot. Additionally, transforming growth factor beta 1 (TGF-β1) inhibitors and TGF-β1 were used to determine whether glucose-regulated Matrilin-2 expression was mediated by the TGF-β1/Smad3 signaling pathway. Our data demonstrated that Matrilin-2 expression was markedly induced by high glucose and TGF-β1. High glucose-induced Matrilin-2 expression was inhibited by TGF-β1/Smad3 inhibitors, indicating that Matrilin-2 was markedly induced by high glucose and this induction was mediated by the TGF-β1/Smad3 pathway. Taken together, our results showed that high-glucose-induced Matrilin-2 expression that was mediated by the TGF-β1/Smad3 signaling pathway might play a role in Diabetic nephropathy (DN) pathogenesis and our finding provided a potential diagnostic and/or therapeutic target for DN.
Collapse
Affiliation(s)
- Shukun Zhang
- Department of Pathology, Qinghai Provincial People's Hospital, 2 Gonghe Road, Xining, Qinghai 810007, China
| | - Menglan Zhang
- Department of Pathology, Qinghai Provincial People's Hospital, 2 Gonghe Road, Xining, Qinghai 810007, China
| | - Hong Huang
- Medical College of Qinghai University, 16 Kunlun Road, Xining 810000, China
| | - Shiying Zhou
- Department of Pathology, Qinghai Provincial People's Hospital, 2 Gonghe Road, Xining, Qinghai 810007, China
| | - Yanshneg Du
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Xin Yi
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Junming Luo
- Department of Pathology, Qinghai Provincial People's Hospital, 2 Gonghe Road, Xining, Qinghai 810007, China.
| |
Collapse
|