REFERENCES

1. Witwer KW, Goberdhan DC, O’Driscoll L, et al. Updating MISEV: evolving the minimal requirements for studies of extracellular vesicles. J Extracell Vesicles 2021;10:e12182.

2. Witwer KW, Théry C. Extracellular vesicles or exosomes? J Extracell Vesicles 2019;8:1648167.

3. Théry C, Witwer KW, Aikawa E, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement Of The International Society For Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles 2018;7:1535750.

4. Liu H, Zhang Q, Wang S, et al. Bacterial extracellular vesicles as bioactive nanocarriers for drug delivery: advances and perspectives. Bioact Mater 2022;14:169-81.

5. O’Brien K, Breyne K, Ughetto S, Laurent LC, Breakefield XO. RNA delivery by extracellular vesicles in mammalian cells and its applications. Nat Rev Mol Cell Biol 2020;21:585-606.

6. Herrmann IK, Wood MJA, Fuhrmann G. Extracellular vesicles as a next-generation drug delivery platform. Nat Nanotechnol 2021;16:748-59.

7. Liu C, He D, Cen H, et al. Nucleic acid functionalized extracellular vesicles as promising therapeutic systems for nanomedicine. EVCNA 2022; doi: 10.20517/evcna.2021.21.

8. Duan L, Xu L, Xu X, et al. Exosome-mediated delivery of gene vectors for gene therapy. Nanoscale 2021;13:1387-97.

9. Li M, Zhou H, Yang C, et al. Bacterial outer membrane vesicles as a platform for biomedical applications: an update. J Control Release 2020;323:253-68.

10. Sharma S, Masud MK, Kaneti YV, et al. Extracellular vesicle nanoarchitectonics for novel drug delivery applications. Small 2021;17:e2102220.

11. Jiang C, Fu Y, Liu G, et al. Multiplexed profiling of extracellular vesicles for biomarker development. Nanomicro Lett 2021;14:3.

12. Tian T, Zhang HX, He CP, et al. Surface functionalized exosomes as targeted drug delivery vehicles for cerebral ischemia therapy. Biomaterials 2018;150:137-49.

13. Xu M, Feng T, Liu B, et al. Engineered exosomes: desirable target-tracking characteristics for cerebrovascular and neurodegenerative disease therapies. Theranostics 2021;11:8926-44.

14. Liao Z, Liu H, Ma L, et al. Engineering extracellular vesicles restore the impaired cellular uptake and attenuate intervertebral disc degeneration. ACS Nano 2021;15:14709-24.

15. Gujrati V, Prakash J, Malekzadeh-Najafabadi J, et al. Bioengineered bacterial vesicles as biological nano-heaters for optoacoustic imaging. Nat Commun 2019;10:1114.

16. Tran PHL, Xiang D, Tran TTD, et al. Exosomes and nanoengineering: a match made for precision therapeutics. Adv Mater 2020;32:e1904040.

17. Song H, Li X, Zhao Z, et al. Reversal of osteoporotic activity by endothelial cell-secreted bone targeting and biocompatible exosomes. Nano Lett 2019;19:3040-8.

18. Hu Y, Li X, Zhang Q, et al. Exosome-guided bone targeted delivery of Antagomir-188 as an anabolic therapy for bone loss. Bioact Mater 2021;6:2905-13.

19. Kim OY, Park HT, Dinh NTH, et al. Bacterial outer membrane vesicles suppress tumor by interferon-γ-mediated antitumor response. Nat Commun 2017;8:626.

20. Gujrati V, Kim S, Kim SH, et al. Bioengineered bacterial outer membrane vesicles as cell-specific drug-delivery vehicles for cancer therapy. ACS Nano 2014;8:1525-37.

21. Kalluri R, LeBleu VS. function, and biomedical applications of exosomes. Science 2020;367:eaau6977.

22. Kim J, Song Y, Park CH, Choi C. Platform technologies and human cell lines for the production of therapeutic exosomes. EVCNA 2021; doi: 10.20517/evcna.2020.01.

23. Work E, Knox KW, Vesk M. The chemistry and electron microscopy of an extracellular lipopolysaccharide from escherichia coli. Ann N Y Acad Sci 1966;133:438-49.

24. Knox KW, Vesk M, Work E. Relation between excreted lipopolysaccharide complexes and surface structures of a lysine-limited culture of Escherichia coli. J Bacteriol 1966;92:1206-17.

25. Schwechheimer C, Kuehn MJ. Outer-membrane vesicles from Gram-negative bacteria: biogenesis and functions. Nat Rev Microbiol 2015;13:605-19.

26. Brown L, Wolf JM, Prados-Rosales R, Casadevall A. Through the wall: extracellular vesicles in Gram-positive bacteria, mycobacteria and fungi. Nat Rev Microbiol 2015;13:620-30.

27. Harding C, Heuser J, Stahl P. Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes. J Cell Biol 1983;97:329-39.

28. Pan B, Johnstone RM. Fate of the transferrin receptor during maturation of sheep reticulocytes. in vitro ;33:967-78.

29. Vidal M. Exosomes: Revisiting their role as “garbage bags”. Traffic 2019;20:815-28.

30. Raposo G, Nijman HW, Stoorvogel W, et al. B lymphocytes secrete antigen-presenting vesicles. J Exp Med 1996;183:1161-72.

31. Prangishvili D, Holz I, Stieger E, et al. Sulfolobicins, specific proteinaceous toxins produced by strains of the extremely thermophilic archaeal genus Sulfolobus. J Bacteriol 2000;182:2985-8.

32. Valadi H, Ekström K, Bossios A, et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol 2007;9:654-9.

33. Fonseka P, Marzan AL, Mathivanan S. Introduction to the Community of Extracellular Vesicles. Subcell Biochem 2021;97:3-18.

34. Mathieu M, Martin-Jaular L, Lavieu G, Théry C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol 2019;21:9-17.

35. Kowal J, Arras G, Colombo M, et al. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc Natl Acad Sci U S A 2016;113:E968-77.

36. Kowal J, Tkach M, Théry C. Biogenesis and secretion of exosomes. Curr Opin Cell Biol 2014;29:116-25.

37. Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol 2013;200:373-83.

38. Trajkovic K, Hsu C, Chiantia S, et al. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science 2008;319:1244-7.

39. Palomino RA, Vanpouille C, Costantini PE, Margolis L. Microbiota-host communications: Bacterial extracellular vesicles as a common language. PLoS Pathog 2021;17:e1009508.

40. Wang S, Gao J, Wang Z. Outer membrane vesicles for vaccination and targeted drug delivery. Wiley Interdiscip Rev Nanomed Nanobiotechnol 2019;11:e1523.

41. Garcia-Martin R, Wang G, Brandão BB, et al. MicroRNA sequence codes for small extracellular vesicle release and cellular retention. Nature 2022;601:446-51.

42. Díaz-Garrido N, Badia J, Baldomà L. Microbiota-derived extracellular vesicles in interkingdom communication in the gut. J Extracell Vesicles 2021;10:e12161.

43. Bittel M, Reichert P, Sarfati I, et al. Visualizing transfer of microbial biomolecules by outer membrane vesicles in microbe-host-communication. in vivo ;10:e12159.

44. Vanaja SK, Russo AJ, Behl B, et al. Bacterial outer membrane vesicles mediate cytosolic localization of LPS and caspase-11 activation. Cell 2016;165:1106-19.

45. Deo P, Chow SH, Han ML, et al. Mitochondrial dysfunction caused by outer membrane vesicles from gram-negative bacteria activates intrinsic apoptosis and inflammation. Nat Microbiol 2020;5:1418-27.

46. Somerville JE Jr, Cassiano L, Darveau RP. Escherichia coli msbB gene as a virulence factor and a therapeutic target. Infect Immun 1999;67:6583-90.

47. Grozdanov L, Raasch C, Schulze J, et al. Analysis of the genome structure of the nonpathogenic probiotic escherichia coli strain nissle 1917. J Bacteriol 2004;186:5432-41.

48. Yan X, Liu XY, Zhang D, et al. Construction of a sustainable 3-hydroxybutyrate-producing probiotic escherichia coli for treatment of colitis. Cell Mol Immunol 2021;18:2344-57.

49. Praveschotinunt P, Duraj-Thatte AM, Gelfat I, et al. Engineered E. coli Nissle 1917 for the delivery of matrix-tethered therapeutic domains to the gut. Nat Commun 2019;10:5580.

50. Domingues S, Nielsen KM. Membrane vesicles and horizontal gene transfer in prokaryotes. Curr Opin Microbiol 2017;38:16-21.

51. Gilmore WJ, Johnston EL, Zavan L, Bitto NJ, Kaparakis-Liaskos M. Immunomodulatory roles and novel applications of bacterial membrane vesicles. Mol Immunol 2021;134:72-85.

52. Lee HJ. Microbe-host communication by small RNAs in extracellular vesicles: vehicles for transkingdom RNA transportation. Int J Mol Sci 2019;20:1487.

53. Dauros-Singorenko P, Blenkiron C, Phillips A, Swift S. The functional RNA cargo of bacterial membrane vesicles. FEMS Microbiol Lett 2018:365.

54. Kumar SR, Kimchi ET, Manjunath Y, et al. RNA cargos in extracellular vesicles derived from blood serum in pancreas associated conditions. Sci Rep 2020;10:2800.

55. la Torre Gomez C, Goreham RV, Bech Serra JJ, Nann T, Kussmann M. “Exosomics”-a review of biophysics, biology and biochemistry of exosomes with a focus on human breast milk. Front Genet 2018;9:92.

56. Zhu Q, Cheng L, Deng C, et al. The genetic source tracking of human urinary exosomes. Proc Natl Acad Sci U S A 2021;118:e2108876118.

57. Liu H, Yuan W, Pang Q, Xue C, Yan X. Single-particle analysis of tear fluid reveals abundant presence of tissue factor-exposing extracellular vesicles with strong coagulation activity. Talanta 2022;239:123089.

58. Lin Y, Dong H, Deng W, et al. Evaluation of salivary exosomal chimeric. GOLM1-NAA35 ;25:3035-45.

59. Höög JL, Lötvall J. Diversity of extracellular vesicles in human ejaculates revealed by cryo-electron microscopy. J Extracell Vesicles 2015;4:28680.

60. Han X, Lee A, Huang S, Gao J, Spence JR, Owyang C. Lactobacillus rhamnosus GG prevents epithelial barrier dysfunction induced by interferon-gamma and fecal supernatants from irritable bowel syndrome patients in human intestinal enteroids and colonoids. Gut Microbes 2019;10:59-76.

61. Liu JH, Chen CY, Liu ZZ, et al. Extracellular vesicles from child gut microbiota enter into bone to preserve bone mass and strength. Adv Sci (Weinh) 2021;8:2004831.

62. Maeki M, Kimura N, Sato Y, Harashima H, Tokeshi M. Advances in microfluidics for lipid nanoparticles and extracellular vesicles and applications in drug delivery systems. Adv Drug Deliv Rev 2018;128:84-100.

63. Zhu L, Xu N, Zhang ZL, Zhang TC. Cell derived extracellular vesicles: from isolation to functionalization and biomedical applications. Biomater Sci 2019;7:3552-65.

64. Hartjes TA, Mytnyk S, Jenster GW, van Steijn V, van Royen ME. Extracellular vesicle quantification and characterization: common methods and emerging approaches. Bioengineering (Basel) 2019;6:7.

65. Li X, Corbett AL, Taatizadeh E, et al. Challenges and opportunities in exosome research-perspectives from biology, engineering, and cancer therapy. APL Bioeng 2019;3:011503.

66. Liang Y, Duan L, Lu J, Xia J. Engineering exosomes for targeted drug delivery. Theranostics 2021;11:3183-95.

67. Li P, Kaslan M, Lee SH, Yao J, Gao Z. Progress in exosome isolation techniques. Theranostics 2017;7:789-804.

68. Gardiner C, Di Vizio D, Sahoo S, et al. Techniques used for the isolation and characterization of extracellular vesicles: results of a worldwide survey. J Extracell Vesicles 2016;5:32945.

69. Xiong Y, Chen L, Yan C, et al. Circulating exosomal miR-20b-5p inhibition restores Wnt9b signaling and reverses diabetes-associated impaired wound healing. Small 2020;16:e1904044.

70. Jiang S, Tian G, Yang Z, et al. Enhancement of acellular cartilage matrix scaffold by Wharton’s jelly mesenchymal stem cell-derived exosomes to promote osteochondral regeneration. Bioact Mater 2021;6:2711-28.

71. Xu X, Liang Y, Li X, et al. Exosome-mediated delivery of kartogenin for chondrogenesis of synovial fluid-derived mesenchymal stem cells and cartilage regeneration. Biomaterials 2021;269:120539.

72. Prados-Rosales R, Brown L, Casadevall A, Montalvo-Quirós S, Luque-Garcia JL. Isolation and identification of membrane vesicle-associated proteins in Gram-positive bacteria and mycobacteria. MethodsX 2014;1:124-9.

73. Chutkan H, Macdonald I, Manning A, Kuehn MJ. Quantitative and qualitative preparations of bacterial outer membrane vesicles. Methods Mol Biol 2013; 966:259-72.

74. Klimentová J, Stulík J. Methods of isolation and purification of outer membrane vesicles from gram-negative bacteria. Microbiol Res 2015;170:1-9.

75. Kim W, Lee EJ, Bae IH, et al.

76. Wang X, Zhang H, Yang H, et al. Cell-derived exosomes as promising carriers for drug delivery and targeted therapy. Curr Cancer Drug Targets 2018;18:347-54.

77. Collins FS, Varmus H. A new initiative on precision medicine. N Engl J Med 2015;372:793-5.

78. Mathivanan S, Fahner CJ, Reid GE, Simpson RJ. ExoCarta 2012: database of exosomal proteins, RNA and lipids. Nucleic Acids Res 2012;40:D1241-4.

79. Lin Y, Wu J, Gu W, et al. Exosome-liposome hybrid nanoparticles deliver CRISPR/Cas9 system in MSCs. Adv Sci (Weinh) 2018;5:1700611.

80. Chen G, Bai Y, Li Z, et al. Bacterial extracellular vesicle-coated multi-antigenic nanovaccines protect against drug-resistant. Staphylococcus aureus ;10:7131-49.

81. Kojima R, Bojar D, Rizzi G, et al. Designer exosomes produced by implanted cells intracerebrally deliver therapeutic cargo for Parkinson’s disease treatment. Nat Commun 2018;9:1305.

82. Wu P, Zhang B, Ocansey DKW, Xu W, Qian H. Extracellular vesicles: a bright star of nanomedicine. Biomaterials 2021;269:120467.

83. Alvarez-Erviti L, Seow Y, Yin H, et al. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol 2011;29:341-5.

84. Liang G, Zhu Y, Ali DJ, et al. Engineered exosomes for targeted co-delivery of miR-21 inhibitor and chemotherapeutics to reverse drug resistance in colon cancer. J Nanobiotechnology 2020;18:10.

85. Liang Y, Xu X, Li X, et al. Chondrocyte-targeted microRNA delivery by engineered exosomes toward a cell-free osteoarthritis therapy. ACS Appl Mater Interfaces 2020;12:36938-47.

86. Liang G, Kan S, Zhu Y, et al. Engineered exosome-mediated delivery of functionally active miR-26a and its enhanced suppression effect in HepG2 cells. Int J Nanomedicine 2018;13:585-99.

87. Kanuma T, Yamamoto T, Kobiyama K, et al. CD63-mediated antigen delivery into extracellular vesicles via DNA vaccination results in robust CD8+ T cell responses. J Immunol 2017;198:4707-15.

88. Jia G, Han Y, An Y, et al. NRP-1 targeted and cargo-loaded exosomes facilitate simultaneous imaging and therapy of glioma. in vitro ;178:302-16.

89. Qi H, Liu C, Long L, et al. Blood exosomes endowed with magnetic and targeting properties for cancer therapy. ACS Nano 2016;10:3323-33.

90. Yang L, Han D, Zhan Q, et al. Blood TfR+ exosomes separated by a pH-responsive method deliver chemotherapeutics for tumor therapy. Theranostics 2019;9:7680-96.

91. Yang Y, Hong Y, Nam GH, et al. Virus-mimetic fusogenic exosomes for direct delivery of integral membrane proteins to target cell membranes. Adv Mater 2017;29:1605604.

92. Gao X, Li S, Ding F, et al. Rapid detection of exosomal micrornas using virus-mimicking fusogenic vesicles. Angew Chem Int Ed Engl 2019;58:8719-23.

93. Piffoux M, Silva AKA, Wilhelm C, Gazeau F, Tareste D. Modification of extracellular vesicles by fusion with liposomes for the design of personalized biogenic drug delivery systems. ACS Nano 2018;12:6830-42.

94. Fang RH, Kroll AV, Gao W, Zhang L. Cell membrane coating nanotechnology. Adv Mater 2018;30:e1706759.

95. Hu CM, Zhang L, Aryal S, et al. Erythrocyte membrane-camouflaged polymeric nanoparticles as a biomimetic delivery platform. Proc Natl Acad Sci U S A 2011;108:10980-5.

96. Su T, Huang K, Ma H, et al. Platelet-inspired nanocells for targeted heart repair after ischemia/reperfusion injury. Adv Funct Mater 2019;29:1803567.

97. Rao L, Bu LL, Cai B, et al. Cancer cell membrane-coated upconversion nanoprobes for highly specific tumor imaging. Adv Mater 2016;28:3460-6.

98. Wang S, Gao J, Li M, Wang L, Wang Z. A facile approach for development of a vaccine made of bacterial double-layered membrane vesicles (DMVs). Biomaterials 2018;187:28-38.

99. Pore D, Chakrabarti MK. Outer membrane protein A (OmpA) from shigella flexneri 2a: a promising subunit vaccine candidate. Vaccine 2013;31:3644-50.

100. Tian Y, Li S, Song J, et al. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials 2014;35:2383-90.

101. Bai J, Duan J, Liu R, et al. Engineered targeting tLyp-1 exosomes as gene therapy vectors for efficient delivery of siRNA into lung cancer cells. Asian J Pharm Sci 2020;15:461-71.

102. Rountree RB, Mandl SJ, Nachtwey JM, et al. Exosome targeting of tumor antigens expressed by cancer vaccines can improve antigen immunogenicity and therapeutic efficacy. Cancer Res 2011;71:5235-44.

103. Gao X, Ran N, Dong X, et al. Anchor peptide captures, targets, and loads exosomes of diverse origins for diagnostics and therapy. Sci Transl Med 2018;10:eaat0195.

104. Zuo B, Qi H, Lu Z, et al. Alarmin-painted exosomes elicit persistent antitumor immunity in large established tumors in mice. Nat Commun 2020;11:1790.

105. Fan Z, Xiao K, Lin J, Liao Y, Huang X. Functionalized DNA enables programming exosomes/vesicles for tumor imaging and therapy. Small 2019;15:e1903761.

106. Di H, Zeng E, Zhang P, et al. General approach to engineering extracellular vesicles for biomedical analysis. Anal Chem 2019;91:12752-9.

107. Wang J, Li W, Lu Z, et al. The use of RGD-engineered exosomes for enhanced targeting ability and synergistic therapy toward angiogenesis. Nanoscale 2017;9:15598-605.

108. Wang J, Li W, Zhang L, et al. Chemically edited exosomes with dual ligand Purified by microfluidic device for active targeted drug delivery to tumor cells. ACS Appl Mater Interfaces 2017;9:27441-52.

109. Khongkow M, Yata T, Boonrungsiman S, et al. Surface modification of gold nanoparticles with neuron-targeted exosome for enhanced blood-brain barrier penetration. Sci Rep 2019;9:8278.

110. Kim MS, Haney MJ, Zhao Y, et al. Engineering macrophage-derived exosomes for targeted paclitaxel delivery to pulmonary metastases:. in vitro ;14:195-204.

111. Cao Y, Wu T, Zhang K, et al. Engineered exosome-mediated near-infrared-II region V2C quantum dot delivery for nucleus-target low-temperature photothermal therapy. ACS Nano 2019;13:1499-510.

112. Pi F, Binzel DW, Lee TJ, et al. Nanoparticle orientation to control RNA loading and ligand display on extracellular vesicles for cancer regression. Nat Nanotechnol 2018;13:82-9.

113. Nakase I, Futaki S. Combined treatment with a pH-sensitive fusogenic peptide and cationic lipids achieves enhanced cytosolic delivery of exosomes. Sci Rep 2015;5:10112.

114. Li Y, Gao Y, Gong C, et al. A33 antibody-functionalized exosomes for targeted delivery of doxorubicin against colorectal cancer. Nanomedicine 2018;14:1973-85.

115. Zhao L, Gu C, Gan Y, et al. Exosome-mediated siRNA delivery to suppress postoperative breast cancer metastasis. J Control Release 2020;318:1-15.

116. Bose RJC, Uday Kumar S, Zeng Y, et al. Tumor cell-derived extracellular vesicle-coated nanocarriers: an efficient theranostic platform for the cancer-specific delivery of Anti-miR-21 and imaging agents. ACS Nano 2018;12:10817-32.

117. Gujrati V, Ntziachristos V. Bioengineered bacterial vesicles for optoacoustics-guided phototherapy. Methods Enzymol 2021;657:349-64.

118. Fu W, Lei C, Liu S, et al. CAR exosomes derived from effector CAR-T cells have potent antitumour effects and low toxicity. Nat Commun 2019;10:4355.

119. Shi X, Cheng Q, Hou T, et al. Genetically engineered cell-derived nanoparticles for targeted breast cancer immunotherapy. Mol Ther 2020;28:536-47.

120. Cui GH, Guo HD, Li H, et al. RVG-modified exosomes derived from mesenchymal stem cells rescue memory deficits by regulating inflammatory responses in a mouse model of Alzheimer’s disease. Immun Ageing 2019;16:10.

121. Cooper JM, Wiklander PB, Nordin JZ, et al. Systemic exosomal siRNA delivery reduced alpha-synuclein aggregates in brains of transgenic mice. Mov Disord 2014;29:1476-85.

122. Liu Y, Li D, Liu Z, et al. Targeted exosome-mediated delivery of opioid receptor Mu siRNA for the treatment of morphine relapse. Sci Rep 2015;5:17543.

123. Kim M, Kim G, Hwang DW, Lee M. Delivery of high mobility group box-1 sirna using brain-targeting exosomes for ischemic stroke therapy. J Biomed Nanotechnol 2019;15:2401-12.

124. Yang J, Zhang X, Chen X, Wang L, Yang G. Exosome mediated delivery of mir-124 promotes neurogenesis after ischemia. Mol Ther Nucleic Acids 2017;7:278-87.

125. Abhange K, Makler A, Wen Y, et al. Small extracellular vesicles in cancer. Bioact Mater 2021;6:3705-43.

126. Zhuang Q, Xu J, Deng D, et al. Bacteria-derived membrane vesicles to advance targeted photothermal tumor ablation. Biomaterials 2021;268:120550.

127. Chronopoulos A, Kalluri R. Emerging role of bacterial extracellular vesicles in cancer. Oncogene 2020;39:6951-60.

128. Huang S, Jiang C, Mughal MJ, Wang G, Xing F. An. in vivo ;10:839078.

129. Han M, Pang B, Zhou C, et al. Liquid biopsy of extracellular vesicle biomarkers for prostate cancer personalized treatment decision. EVCNA 2022; doi: 10.20517/evcna.2021.20.

130. Mulligan RC. The basic science of gene therapy. Science 1993;260:926-32.

131. Vader P, Mol EA, Pasterkamp G, Schiffelers RM. Extracellular vesicles for drug delivery. Adv Drug Deliv Rev 2016;106:148-56.

132. Zheng L, Hu X, Wu H, et al.

133. Webb JA, Ou YC, Faley S, et al. Theranostic gold nanoantennas for simultaneous multiplexed Raman imaging of immunomarkers and photothermal therapy. ACS Omega 2017;2:3583-94.

134. Stritzker J, Kirscher L, Scadeng M, et al. Vaccinia virus-mediated melanin production allows MR and optoacoustic deep tissue imaging and laser-induced thermotherapy of cancer. Proc Natl Acad Sci U S A 2013;110:3316-20.

135. Fan Q, Cheng K, Hu X, et al. Transferring biomarker into molecular probe: melanin nanoparticle as a naturally active platform for multimodality imaging. J Am Chem Soc 2014;136:15185-94.

136. Cheng Q, Shi X, Han M, et al. Reprogramming exosomes as nanoscale controllers of cellular immunity. J Am Chem Soc 2018;140:16413-7.

137. Xiao L, Hareendran S, Loh YP. Function of exosomes in neurological disorders and brain tumors. Extracell Vesicles Circ Nucl Acids 2021;2:55-79.

138. Cuesta CM, Guerri C, Ureña J, Pascual M. Role of microbiota-derived extracellular vesicles in gut-brain communication. Int J Mol Sci 2021;22:4235.

139. Hampel H, Vassar R, De Strooper B, et al. The β-Secretase BACE1 in Alzheimer’s disease. Biol Psychiatry 2021;89:745-56.

140. Jiang C, Hopfner F, Katsikoudi A, et al. Serum neuronal exosomes predict and differentiate Parkinson’s disease from atypical parkinsonism. J Neurol Neurosurg Psychiatry 2020;91:720-9.

141. Jiang C, Hopfner F, Berg D, et al. Validation of α-Synuclein in L1CAM-immunocaptured exosomes as a biomarker for the stratification of parkinsonian syndromes. Mov Disord 2021;36:2663-9.

142. Fu Y, Jiang C, Tofaris GK, Davis JJ. Facile Impedimetric analysis of neuronal exosome markers in Parkinson’s disease diagnostics. Anal Chem 2020;92:13647-51.

143. Powers WJ. Acute ischemic stroke. N Engl J Med 2020;383:252-60.

144. Dharap A, Bowen K, Place R, Li LC, Vemuganti R. Transient focal ischemia induces extensive temporal changes in rat cerebral microRNAome. J Cereb Blood Flow Metab 2009;29:675-87.

145. Jeyaseelan K, Lim KY, Armugam A. MicroRNA expression in the blood and brain of rats subjected to transient focal ischemia by middle cerebral artery occlusion. Stroke 2008;39:959-66.

146. Ren X, Liu H, Wu X, et al. Reactive oxygen species (ROS)-responsive biomaterials for the treatment of bone-related diseases. Front Bioeng Biotechnol 2021;9:820468.

147. Chen S, Chen X, Geng Z, Su J. The horizon of bone organoid: a perspective on construction and application. Bioact Mater 2022;18:15-25.

148. Hu Y, Chen X, Wang S, Jing Y, Su J. Subchondral bone microenvironment in osteoarthritis and pain. Bone Res 2021;9:20.

149. Gu J, Zhang Q, Geng M, et al. Construction of nanofibrous scaffolds with interconnected perfusable microchannel networks for engineering of vascularized bone tissue. Bioact Mater 2021;6:3254-68.

150. Puhm F, Boilard E, Machlus KR. Platelet extracellular vesicles: beyond the blood. Arterioscler Thromb Vasc Biol 2021;41:87-96.

151. Wu X, Wang Y, Xiao Y, et al. Extracellular vesicles: potential role in osteoarthritis regenerative medicine. J Orthop Translat 2020;21:73-80.

152. Liu Y, Ma Y, Zhang J, Yuan Y, Wang J. Exosomes: a novel therapeutic agent for cartilage and bone tissue regeneration. Dose Response 2019;17:1559325819892702.

153. Tao SC, Guo SC. Extracellular vesicles in bone: “dogrobbers” in the “eternal battle field”. Cell Commun Signal 2019;17:6.

154. Zhang Y, Xie Y, Hao Z, et al. Umbilical mesenchymal stem cell-derived exosome-encapsulated hydrogels accelerate bone repair by enhancing angiogenesis. ACS Appl Mater Interfaces 2021;13:18472-87.

155. Re F, Gabusi E, Manferdini C, Russo D, Lisignoli G. Bone regeneration improves with mesenchymal stem cell derived extracellular vesicles (EVs) combined with scaffolds: a systematic review. Biology (Basel) 2021;10:579.

156. Pishavar E, Luo H, Naserifar M, et al. Advanced hydrogels as exosome delivery systems for osteogenic differentiation of MSCs: application in bone regeneration. Int J Mol Sci 2021;22:6203.

157. Fan J, Lee CS, Kim S, et al. Generation of small rna-modulated exosome mimetics for bone regeneration. ACS Nano 2020;14:11973-84.

158. Zha Y, Li Y, Lin T, et al. Progenitor cell-derived exosomes endowed with VEGF plasmids enhance osteogenic induction and vascular remodeling in large segmental bone defects. Theranostics 2021;11:397-409.

159. Zhai M, Zhu Y, Yang M, Mao C. Human mesenchymal stem cell derived exosomes enhance cell-free bone regeneration by altering their miRNAs profiles. Adv Sci (Weinh) 2020;7:2001334.

160. Lazar S, Mor S, Chen J, Hao D, Wang A. Bioengineered extracellular vesicle-loaded bioscaffolds for therapeutic applications in regenerative medicine. Extracell Vesicles Circ Nucl Acids 2021;2:175-8.

161. Xue S, Zhou X, Sang W, et al. Cartilage-targeting peptide-modified dual-drug delivery nanoplatform with NIR laser response for osteoarthritis therapy. Bioact Mater 2021;6:2372-89.

162. Glyn-jones S, Palmer AJR, Agricola R, et al. Osteoarthritis. The Lancet 2015;386:376-87.

163. Martel-Pelletier J, Barr AJ, Cicuttini FM, et al. Osteoarthritis. Nat Rev Dis Primers 2016;2:16072.

164. Ragni E, Orfei CP, Papait A, de Girolamo L. Comparison of miRNA cargo in human adipose-tissue vs. amniotic-membrane derived mesenchymal stromal cells extracellular vesicles for osteoarthritis treatment. EVCNA 2021; doi: 10.20517/evcna.2021.11.

165. Johnson K, Zhu S, Tremblay MS, et al. A stem cell-based approach to cartilage repair. Science 2012;336:717-21.

166. Kang ML, Ko JY, Kim JE, Im GI. Intra-articular delivery of kartogenin-conjugated chitosan nano/microparticles for cartilage regeneration. Biomaterials 2014;35:9984-94.

167. Xu X, Shi D, Shen Y, et al. Full-thickness cartilage defects are repaired via a microfracture technique and intraarticular injection of the small-molecule compound kartogenin. Arthritis Res Ther 2015;17:20.

168. Zhang R, Ma J, Yao J. Molecular mechanisms of the cartilage-specific microRNA-140 in osteoarthritis. Inflamm Res 2013;62:871-7.

169. Miyaki S, Sato T, Inoue A, et al. MicroRNA-140 plays dual roles in both cartilage development and homeostasis. Genes Dev 2010;24:1173-85.

170. Black DM, Rosen CJ. Clinical Practice. Postmenopausal osteoporosis. N Engl J Med 2016;374:254-62.

171. Li X, Wang L, Huang B, et al. Targeting actin-bundling protein L-plastin as an anabolic therapy for bone loss. Sci Adv 2020;6:eabb7135.

172. Zhao H, Li X, Zhang D, et al. Integrative bone metabolomics-lipidomics strategy for pathological mechanism of postmenopausal osteoporosis mouse model. Sci Rep 2018;8:16456.

173. Brown C. Osteoporosis: staying strong. Nature 2017;550:S15-7.

174. Li CJ, Cheng P, Liang MK, et al. MicroRNA-188 regulates age-related switch between osteoblast and adipocyte differentiation. J Clin Invest 2015;125:1509-22.

175. Gujrati V, Lee M, Ko YJ, et al. Bioengineered yeast-derived vacuoles with enhanced tissue-penetrating ability for targeted cancer therapy. Proc Natl Acad Sci U S A 2016;113:710-5.

176. MacDiarmid JA, Mugridge NB, Weiss JC, et al. Bacterially derived 400 nm particles for encapsulation and cancer cell targeting of chemotherapeutics. Cancer Cell 2007;11:431-45.

177. Gudbergsson JM, Jønsson K, Simonsen JB, Johnsen KB. Systematic review of targeted extracellular vesicles for drug delivery - considerations on methodological and biological heterogeneity. J Control Release 2019;306:108-20.

178. Xue X, Hu Y, Deng Y, Su J. Recent advances in design of functional biocompatible hydrogels for bone tissue engineering. Adv Funct Mater 2021;31:2009432.

179. Chen W, Zhou Z, Chen D, et al. Bone regeneration using MMP-cleavable peptides-based hydrogels. Gels 2021;7:199.

180. Xiong Y, Chen L, Liu P, et al. All-in-one: multifunctional hydrogel accelerates oxidative diabetic wound healing through timed-release of exosome and fibroblast growth factor. Small 2022;18:e2104229.

181. Li L, Yu F, Zheng L, et al. Natural hydrogels for cartilage regeneration: Modification, preparation and application. J Orthop Translat 2019;17:26-41.

182. Zou Y, Huang B, Cao L, Deng Y, Su J. Tailored mesoporous inorganic biomaterials: assembly, functionalization, and drug delivery engineering. Adv Mater 2021;33:e2005215.

183. Geng Z, Sang S, Wang S, et al. Optimizing the strontium content to achieve an ideal osseointegration through balancing apatite-forming ability and osteogenic activity. Mater Sci Eng C Mater Biol Appl 2022:112647.

184. Zipkin M. Big pharma buys into exosomes for drug delivery. Nat Biotechnol 2020;38:1226-8.

185. Liu H, Wang Y, Hou Y, Li Z. Fitness of chassis cells and metabolic pathways for l-cysteine overproduction in escherichia coli. J Agric Food Chem 2020;68:14928-37.

186. Liu H, Hou Y, Wang Y, Li Z. Enhancement of sulfur conversion rate in the production of l-cysteine by engineered escherichia coli. J Agric Food Chem 2020;68:250-7.

187. Yang D, Park SY, Park YS, Eun H, Lee SY. Metabolic engineering of escherichia coli for natural product biosynthesis. Trends Biotechnol 2020;38:745-65.

188. Choi KR, Jang WD, Yang D, et al. Systems metabolic engineering strategies: integrating systems and synthetic biology with metabolic engineering. Trends Biotechnol 2019;37:817-37.

189. Service RF. Synthetic biology. Synthetic biologists design ‘living materials’ that build themselves. Science 2014;343:1421.

190. Tang T, An B, Huang Y, et al. Materials design by synthetic biology. Nat Rev Mater 2021;6:332-50.

191. Depommier C, Everard A, Druart C, et al. Supplementation with akkermansia muciniphila in overweight and obese human volunteers: a proof-of-concept exploratory study. Nat Med 2019;25:1096-103.

192. Riehl TE, Alvarado D, Ee X, et al. Lactobacillus rhamnosus GG protects the intestinal epithelium from radiation injury through release of lipoteichoic acid, macrophage activation and the migration of mesenchymal stem cells. Gut 2019;68:1003-13.

193. Ozdemir T, Fedorec AJH, Danino T, Barnes CP. Synthetic biology and engineered live biotherapeutics: toward increasing system complexity. Cell Syst 2018;7:5-16.

194. Charbonneau MR, Isabella VM, Li N, Kurtz CB. Developing a new class of engineered live bacterial therapeutics to treat human diseases. Nat Commun 2020;11:1738.

Extracellular Vesicles and Circulating Nucleic Acids
ISSN 2767-6641 (Online)
Follow Us

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/