Anda di halaman 1dari 37

JOURNAL READING JARINGAN KOMPLEKS DARI BEBERAPA FAKTOR DALAM PATOGENESIS LEIOMIOMA UTERI

KEPANITERAAN KLINIK ILMU KEBIDANAN DAN KANDUNGAN

FAKULTAS KEDOKTERAN UKRIDA RUMAH SAKIT UMUM DAERAH TARAKAN

Nama Mahasiswa NIM Nama Pembimbing

: Mohd Asrul B. Che Rahim : 112012059 : Dr Harianto Wijaya, Sp.OG

Jaringan yang Kompleks dari beberapa faktor dalam Patogenesis Leiomioma Uteri Pendahuluan Leiomioma uteri(fibroids atau myomas) adalah satu-satunya tumor jinak uterus yang paling sering diindikasikan untuk di operasi hysterectomy. Leiomioma uteri secara klinikal timbul

pada 25% dari wanita dalam umur reproduktif dan menyebabkan morbiti seperti perdarahaan berat atau memanjang pada menstruasi,nyeri pada pelvic dan disfungsi reproduksi. Tatalaksana yang definitif untuk tumor fibroid adalah operasi, dan beberapa teknik invasif yang minimal. Estrogen dan Progesteron merupakan promoter pertumbuhan fibroid. Namun growth factor, cytokines dan chemokines juga diduga effektor yang potensial terhadap estrogen dan progestron. Tambahan pula perubahan genetik,mekanisme epigenetic dan komponen matriks extraselular diduga penting dalam inisiasi dan perumbuhan tumor ini.

Tumor uterus fibroid dan prevelensi ras. Prevelensi leimioma uteri adalah 3 kali lipat pada wanita berkulit hitam dari wanita berkulit putih.Faktor lain seperti menarche yang awal,umur(usia reproduktif yang

telat),keturunan,nulliparitas,obesitas,sindrom polikistik ovari,diabetis,hipertensi dan konsumsi alkohol mempunyai faktor meningkatkan risiko berkembangnya leiomioma uteri. Faktor genetik Leiomioma uteri adalah tumor yang indipenden dan monoclonal;pertumbuhan tumor berasal dari 1 sel miometrial yang mutasi. Defek keturunan dari gene fumarate hydratase (FH),Birt-HoggDube (BHD) dan tuberous sclerosis complex 2 akan mengkontribusi terhadap perkembangan tumor ini.Penelitian menunjukkan perbedaan pada perubahan genetik pada leimioma seperti delesi kromosom trisomy, translokasi kromosom dan monosomi kromosom Tambahan El Gharib et al .

menemukan kelainan kromosom klonal pada lima kromosom telah mengidentifikasi tiga loci pada

berbeda (2,7,8,12 dan 22) . Baru-baru ini Cha et al

kromosom 10q24.33,22q13.1, dan 11p15.5 yang berasosiasi dengan kerentanan terhadap uterine fibroid. Dengan konsistensi alterasi genetik kromosom dengan tempat yang tertentu,beberapa kandidat gene telah diidentifikasi HMGA2 sering diekspresi pada leiomioma uteri, dengan penyusunan kembali kromosom 12q15 ,yang mana target gene nya let -7 famili (microRNAs) . Tambahan pula RAD1L1, . Baru-baru ini ekspresi

(miRNAs) dan ditemukan disupresi oleh let-7 secara in-vitro adalah kandidat pasangan translokasi HMGA2 pada leiomioma

mRNA HMGA1 lebih tinggi telah diobservasi berbanding dengan tumor normal secara sitogenetik dengan alterasi 6p21 . Beberapa gene kandidat, seperti PCOLCE,ORC5L dan

LHFPL3,telah dipetakan pada interval kritikal pada kromosom 7q22

. Baru baru ini

penjelasan secara komprehensif terhadap alterasi genetik (mutasi MED12) pada leiomioma uteri telah dilaporkan oleh berbeda-beda kumpulan 45,46,47,48,49,50. Mediator Complex subunit 12 (MED12) adalah komponen mediator kompleks yang mempunyai peran regulator pada aktiviti RNA polimerase II. MED12 mempunyai prean pada aktivasi transkripsi dab represi dan terlibat pada banyak proses pekembangan. Makinen dan teman-teman menemukan bahwa gene MED12 telah bermutasi pada frekuensi tinggi (70%) pada leiomioma uteri didapat pada pasien yang berasal dari Finland (Caucasian). Semua mutasi ini berkluster pada exon 2, dengan lapan mutasi yang men codon 44. Pola yang sama mutasi MED12 exon 2 juga ditemukan pada pasien berasal dari Afrika Selatan. Baru-baru ini Je et al. menemukan studi tentang MED 12 pada 1862 sampel termasuk

leiomioma,pelbagai carcinoma leukemia sarkoma dan tumor stromal lain-lain,yang mana ditemukan kadar frekuensi tinggi MED 12 tetapi hanya 1 pada tumor maligna. Temuan ini dan temuan sebelumnya menyarankan bahwa mutasi MED12 exon 2 mungkin tisu

spesifik terhadap leiomioma uteri.Namun baru-baru ini studi lain merekomendasi kan bahwa mutasi ini tidak hanya pada tumor jinak, pengkaji menemukan bahwa 11% tumor otot yang bepotensi maligna dan 20% leiomiosarkoma uteri ada mutasi MED12. Markawski et al .

. Mendemonstasikan bahwa mutasi MED 12 pada leiomioma uteri

kebanyakan diasosiasikan dengan karyotipe normal tumor. Kumpulan ini juga menemukan bahwa tumor dengan mutasi MED12 mengekspresikan mRNA WNT 4 pada kadar lebih tinggi,menunjukkan bahwa mutasi MED 12 mungkin terlibat dengan aktivasi jalur Wnt. Namun hasil imunohistokimia menunjukkan bahwa tidak ada asosiasi antara status MED 12 dan B-catenin nuclear/ lokalisasi sitoplasmik

Faktor epigenetik Epigenetik adalah semua perubahan diturunkan dalam ekpresi gen yang tidak dikodekan dalam urutan DNA DNA Methylation Deoxyribonucleci acid methylation berlaku pada karbon ke -5 cytosine,Metilasi ini berlaku dipelbagai proses perkembangan dengan menghilangkan,mengalih dan menstabilkan

gen.Ketidakseimbangan ekspresi DNA Metil transferase dijumpai pada leimioma uteri berbanding di miometrium yang sehat.

Modifikasi histone Modifikasi histone adalah faktor epigenetik kedua terpenting dalam peran meregulasi ekspresi gene. Protein histone bisa dimodifikasi dalam pelbagai cara pada N terminal,termasuk asetilasi,fosforilasi metilasi,deaminasi isomerisasi proline dan adenosinosine difosfat ribosilasi. Histone deasetilasi 6(HDAC6) adalah faktor regulator dalam jaringan pengangkutan endokrin dan mempunyai aktiviti deasetilasi histone. Menurut penelitian Wei et al. menguji

ekspresi HDCA6 dan peran patologi nya terhadap leiomioma uteri. Mereka menemukan pola peningkatan HDAC6 dan ekspresi ER- dalam sampel leiomioma. Dapat dikenalpasti bahawa modifikasi epigenetik diperlukan semasa perkembangan dan memainkan peran penting dalam diferensiasi selular dan tissue normal perkembangan menjadi dewasa. Namun sewaktu stadium penting dalam perkembangan,eksposur lingkungan boleh merubah keadaan genom yang terkait dengan program deferensiasi sel atau organ,dengan itu

mengalakkan kerentanan terhadap penyakit ini diwaktu kemudian kehidupan. Penelitian melaporkan melalui efek nongenomik terhadap program ulang pembentukan uterus,estrogen dari linkungan merekrut regulator epigenetik EZH2 dan mengurangkan kadar histone represif dalam kromatin dan mempromosikan tumorigenesis di uteri. Micro RNA Micro RNA adalah non protein coding yang meregulasi banyak proses biologi dengan merencanakan miRNA untuk pembelahan atau represi translasi . Penelitian menunjukkan

beberapa mRNA termasuk let7,miR-21,miR-93,miR 106b dan miR 200 dan target gene mereka secara signifikan di disregulasi pada leiomioma uteri berbanding miometrium normal. Pan et al. melaporkan bahawa miR-21 di ekspresikan berlebihan pada leiomioma, dengan peningkatan spesifik waktu fase sekretorik pada siklus menstruasi pada wanita yang menggunakan kontrasepsi oral. Zavadil et al meneliti korelasi pola global antara ekspresi miRNA yang diubah dengan predicted target genes pada leiomioma uteri dan miometria. Mereka menemukan jumlah miRNA didisregulasi secara korelasi terbalik dengan targetnya pada tingkat protein. Pola asosiasi terbalik miRNA dengan ekspresi pelbagai kandidat mRNA pada leiomioma uteri menunjukkan penglibatan

jalur,termasuk jalur extensive transcriptional

reprogramming,cell

proliferation control,mitogen activated protein kinase MAPK transforming growth factor (TGF),WNT, Janus Kinase/signal trasnduser dan activator of transcription signalling, remodelling perlekatan sel dan sel-sel dan sel-matrix contacts. Estrogen Estrogen mengeluarkan efek fisiologis terhadap target sel nya dengan mengikat ke Estrogen receptor (ER-)dan Estrogen receptor (ER-). Estrogen dan reseptornya memainkan peran

penting dalam fisiologi miometrium dan pertumbuhan leiomioma uteri. Beberapa kumpulan peneliti menemukan mRNA dan protein expression level of ER ( diekspresi lebih tinggi) dan ER- adalah lebih tinggi pada leiomima uteri berbanding miometrium normal. Penelitian terbaru menunjukkan estrogen boleh mempertahankan kadar Progestron receptor (PR) dan dengan demikian progestron melalui reseptornya dapat menggalakkan pertumbuhan leiomioma.

Estrogen secara signifikan mengurangkan ekspresi p53 ( protein supresor tumor). Estrogen dapat meransang proliferasi sel leiomioma dengan meregulasi ekspresi faktor pertumbuhan dan dengan mengaktivasi jalur sinyal. Estrogen juga mencetuskan pengaktifan cepat dan sementara jalur MAPK pada sel leiomioma. Estrogen juga menghasilkan fosforilasi protein tyrosine pada protein intraselular

Progesterone Progesteron bekerja dengan berinteraksi dengan reseptor progesterone dan reseptor reseptor (PR) Progesterene dan reseptornya memainkan peran dalam proses biologi leiomyoma dan miometrium. Penelitian menunjukkan kadar progesterone dan mRNA pada leiomioma lebih tinggi dari miometrium. Fujimoto et al. menemukan ekspresi mRNA PR relatif lebih pada permukaan leiomyoma.Ini menunjukkan ekspresi predominan PR-B pada bagian ini adalah fenotip aktif untuk proliferasi progestasi yang berkaitan dengan pertumbuhan leiomioma. Progesterone dapat menstimulasi pertumbuhan sel leiomioma dan ketahanan hidup sel dengan meningkatkan regulasi sel limfoma B ekspresi (Bcl)-2 protein dan menurunkan regulasi ekspresi tumor necrosis factor (TNF)

Growth factors Growth factor adalah protein atau peptida yang diproduksi oleh sel otot polos dan fibroblas yang turut berperan dalam beberapa aktivitas selelur seperti proliferasi, sintesis ECM dan angiogenesis yang penting untuk pertumbuhan leiomioma. Beberapa growth factors seperti EGF,Heparin binding EGF,PDGF,IGF, TGF- ,TGF- vascular endothelial growth factor (VEGF) , acidic firbroblast growth factor(FGF) dan reseptor masing-masing growth factors,dilaporkan memainkan peran dalam pertumbuhan leiomioma. Activin dan miostatin adalah protein yang diindentifikasi pada leiomioma dan miometrium. Sitokin Sitokin adalah protein yang dilepaskan oleh sel sistem imun . Sitokin menghantar sinyal intraselular dengan mengikat specific cell-surface receptor 151,152. Beberapa interleukin (IL) telah diidentifikasi dalam patofisiologi miometrium. Termasuk IL1,IL-, IL 11, dan IL 13,kerna ekspresi berlebihan sitokin berikut bertindak sebagai kunci regulator kepada subepithelial airway fibrosis. Terutama melalui interaksi dengan TGF-

156,157,158,159. Ekspresi sitokin tersebut lebih pada leiomioma berbanding miometrium. ILs menambah ekspresi TNF- yang ditemukan pada leiomioma relatif pada miometrium normal dan diturunkan regulasi oleh progesteron. Chemokines Chemokines adalah protein yang di ekspresi kan oleh hampir semua sel yang bernukleas. Chemokines adalah mediator angiogenesis,hematopoiesis dan fibrosis. Monocyte

chemoattractant protein 1 memainkan peran dalam respon inflamasi monosit dan makrofag.

Sozen et al. menemukan chemokine lebih tinggi pada miometrium berbanding leiomioma. Apabila sel leiomioma diterapi dengan anti-MCP-1 neutralizing antibody,didapati berlaku proliferasi.Tambahan lagi pada sel leiomioma E2 dan progestin sendiri dan pada kombinasi mengurangkan kadar MCP-1. Hasil ini menunjukkan MCP-1 mempunyai sifat antineoplastik pada leiomioma. IL-8 dan reseptor IL-8 tipe A telah diidentifikasi dengan peningkatan ekspresi pada miometrium berbanding leiomioma dan inhibisi proliferasi sel diobservasi apabila IL-8 diblok oleh antibodi neutral,ini menunjukkan potensi peran IL-8 dalam pertumbuhan tisu miometrium. Tambahan pula beberapa chemokines dan reseptor chemokines termasuk macrophage inflammatory protein (MIP)-1, MIP-1, RANTES, eotaxin, eotaxin-2, IL-8, chemokine (cc-motif) receptor 1 (CCR1), CCR3, CCR5, chemokine (cxc-motif) receptor 1 (CXCR1), and CXCR2 mRNA, telah diidentifikasi pada miometrium dan tisu leiomioma,lebih rendah daripada miometrium. Komponen ECM Leiomioma uteri dikarakterisasi oleh kuantiti dan kualitas abnomaliti pada komponen ECM, terutama kolagen, fibronectin, and proteoglycans. Leiomioma mengandungi 50% lebih daripada miometrium bersamaan dan berfungsi sebagai reservoir untuk growth factors,sitokin chemokines angiogenic dan mediator respon inflamasi dan proteases yang diproduksi oleh sel tumor. Kolagen adalah komponen major pada ECM yang berkontribusi kepada stabilisasi dan mempertahankan integriti struktur tisu. Namun struktur dan orientasi fibrin kolagen yang

abnormal ditemukan pada leiomioma. Disamping itu ekpresi relatif berlebihan pada mRNA kolagen tipe I dan III pada leiomioma berbanding miometrium disebelahnya. Tambahan pula peningkatan ekspresi kolagen tipe I dan V pada tingkat protein ditemukan pada leiomioma

berbanding miometrium yang normal.

Malik et al. menguji beberapa seri kolagen subtipe

COL1A1, 4A2, 6A1, 6A2, 7A1, dan 16A1 diekspresikan lebih banyak pada leiomioma berbanding pada miometrium. Leiomioma uteri pada manusia yang berasal dari fibroblas dapat menstimulisasi proliferasi sel leiomioma uteri dengan peningkatan produksi kolagen tipe type I, IGFBP-3, VEGF, EGF, bFGF, PDGF-A and B, TGF-1, and TGF-3 dan dapat juga mengaktifkan reseptor tyrosine kinase dan reseptor sinyal TGF-. Penemuan ini menunjukkan bahawa pertumbuhan leiomioma dapat dimediasi oleh mekanisme autokrin atau parakrin. Tumor yang berasal dari fibroblas dan atau sel leiomioma uteri dapat menggalakkan sintesis growth factors dan mengaktif kan jalur sinyalnya yang penting dalam menstimulasi proliferasi sel tumor dan produksi komponen ECM. Penelitian menunjukkan perubahan pada ECM dapat memodifikasi stress mekanikel pada sel yang menyebabkan aktivasi internal mechanical signaling dan menyumbang pada pertumbuhan leiomioma. Telah dilaporkan sel leiomioma yang terpajan pada beban mekanikal dan menunjukkan fitur struktural dan biokimia yang konsisten dengan aktivasi solid state signalling.Walaupun fitur leiomioma meningkatkan stress mekanikal sel leiomioma menunjukkan respons melemah pada isyarat mekanikal berbanding pada sel miometrium. Berdasarkan persoalan ini, Malik et al. mengkarakterisasi integrin dan laminin kepada sinyal pada sel leimioma.Hasil eksperimen mereka menunjukkan hasil bahwa sinyal mekanikal yang dilemahkan pada sel leiomioma diikuti oleh peningkatan ekspresi dan ketergantungan pada sinyal integrin 1 pada sel leiomioma berbanding daripada sel miometrium. Dermatopontin, protein ekstraselular yang mengikat kepada molekul kecil demantan sulfate dan decorin, diekspresikan pada leiomioma,namum kadar ekspresi berkurang pada leiomioma berbanding daripada miometrium.Mengingat Leiomioma dan keloid adalah penyakit fibrotik dan berkongsi kesamaan epidemiologik, kaitan molekular telah dibina antara kedua kondisi

patologik dengan melihat berkurangnya ekspresi dermatopontin (kolagen pengikat protein) dan struktur fibril kolagen. Disamping itu beberapa analisa mikroarray menunjukkan penurunan regulasi dermtopontin pada leiomioma berbanding dengan miometrium. Proteoglikan adalah protein glikosilasi yang terikat secara kovalen pada glikoaminoglikan sulfat dan bagian penting kepada struktur leiomioma. David et al. menemukan kadar glikoaminglikan dan versican ( proteoglikan ECM besar) yang tinggi pada fibroid uteri,namun kadar dekorin yang relatif rendah pada fibroid uteri dan keloid berbanding tisu normal. Penemuan ini konsisten dengan penemuan sebelumnya yang berkaitan dengan ikatan molekular antara kedua jenis penyakit fibrotik ini. Tambahan pula beberapa penelitian menunjukkan kadar ekspresi versican adalah lebih tinggi pada leiomioma berbanding daripada miometrium. Pelbagai ekspreis

glikosaminoglikan dan proteoglikan juga ditemukan pada leiomioma dan miometrium. Fibronectin adalah glikoprotein ECM yang mengikat kolagen kepada integrin.Terdapat Kadar ekspresi mRNA fibronectin pada pelbagai patofisiologi miometrium. Stewart et al. menemukan tidak ada perbedaan yang signifikan atara leiomioma dan miometrium pada apa pun tahap pada siklus menstruasi.Namun Arici and Sozen menemukan peningkatan kadar ekspresi fibronectin pada leiomioma berbanding daripada autologous miometrium. Menariknya, kumpulan ini turut melaporkan bahwa TGF-3 menginduksi ekspresi fibronectin pada leiomioma dan secara lansung menstimulasi sel miometrium dan leiomioma pada kultur berpoliferasi. Tissue remodelling yang melibatkan ECM turnover memainkan peran penting pada regresi leiomioma,dimana diregulasi oleh kombinasi aksi oleh matriks

pertumbuhan dan

metalloproteinase (MMP) (protein yang memecahkan ECM) dan tissue inhibitor of MMPs (TIMPs). Khususnya beberapa bentuk dari mRNA MMPs dan TIMPs dan protein telah

ditemukan diekspresikan secara berbeda pada miometrium dan leiomioma. Baru-baru ini analisis mikroarray menunjukkan perubahan pada array secara meluas pada MMP pada tisu leiomioma. Sel Kandidat untuk perkembangan dan pertumbuhan fibroid Dengan mempertimbangkan bahwa fibroid uteri adalah sangat lazim dan mempengaruhi wanita pada usia reproduksi (adanya siklus menstruasi) menambahkan lagi kepercayaan bahwa cedera yang berhubungan dengan dengan mens dapat menyebabkan respon inflasmasi yang tidak benar, ini membawa kepada pembentukan fibroid uteri. Kontraksi miometrium pada akhir

perdarahan menstruasi dapat menginduksi cedera iskemi atau cedera iskemi-reperfusi pada otot polos miometrium yang mungkin menjadi kandidat untuk sel progenitor dari fibroid uteri. Untuk mendukung hipotesis ini, sel apoptotik positif , sel positif p53 dan sel positif 21 telah ditemukan hanya pada fase folikuler pada siklus menstruasi. Namum sel positif Ki-67 yang dilihat terutama pada fase luteal siklus menstruasi. Hasil ini memberi kesan bahwa majoriti dari sel yang rusak kelihatan dieliminasi pada fase folikuler siklus menstruasi, dieliminasi sebagai sel apoptotik , tetapi beberapa sel yang rusak bisa bertahan hidup dengan menerima mekanisme pertahanan terhadap stress oxidatif dan apoptosis. Sel ini mungkin adalah progenitor kepada fibroid uteri. Bahkan sel leiomioma uteri menpunyai mekanisme proteksi terhadap stress mekanisme yang mengekspresi manganase superoxide dismutase (MnSOD) dan terhadap apoptosi Bcl-2, PEP-19(protein sel Purkinje 4 PCP4) dan secreted frizzled related protein1 (sFRP1) Telah dipersetujui bahwa menstruasi ovulasi dan parturitas menyebabkan kecederaan fisiologi yang mencetuskan reaksi inflamasi pada uterus. Sel Miofibroblas diaktifkan oleh inflamasi dan transformasi selular inilah yang menjadi peristiwa penting untuk pemulihan homeostasis tisu dan

proses

penyembuhan

luka.

Miofibroblas

dikarekterisasi

oleh

pertambahan

proliferasi,kebolehan bermigrasi, produksi sitokin dan kapasitas untuk menghasilkan matriks interstitial. Namun fungsi miofiblas yang tidak tepat telah ditunjukkan dapat menyebabkan fibrosis,oleh kerana memiliki sifat ketidakmampuan regenerasi tisu,selalu menghasilkan kolagen kolagen dan parut kaku.. Transformasi miofibroblas dapat berlaku dari sel berbeda tipe,kerna miometrium mengandungi sel otot polos , jaringan ikat fibroblas,stem sel,sel vascular dan progenitor sel berasal dari sumsum tulang. Baru- baru ini penelitian mengenai stem sel telah membuka kemungkinan baru untuk memahami pertumbuhan uterine fibroid. Kesimpulan dan perspektif masa hadapan. Uterine fibroid umumnya berhubungan dengan perdarahan menstruasi yang memanjang, berat dan nyeri pelvis. Namun tumor ini juga mempunyai efek negatif terhadap fertilitas dan hasil kehamilan untuk pasien yang ingin mengikuti assisted reproductive technology (ART). Namun literatur berkaitan dengan leiomioma uteri dan kesan nya kepada ART adalah membingungkan. Tambahan pula prevelensi tinggi leiomioma uteri dan pengaruh potensinya yang merugikan terhadap ART dan fungsi reproduksi. Ini menjamin penelitian yang dirancang teliti diteruskan untuk memastikan etiologi,terapi optimal dan terapi baru yang kurang morbiditasnya. Perbedaan ras dan faktor risiko lain,faktor genetik,mekanisme epigenetik, estrogen, progesteron, growth factors,sitokin,chemokines. Dan komponen ECM telah dikenalpasti memberi kesan kepada biologi miometium dan leiomioma. Beberapa dari faktor tersebut boleh dipertimbangkan untuk membina strategi terapi untuk menghalang transformasi miometium dan/ atau

pertumbuhan miometrium. Sehingga sekarang terapi yang mungkin telah diuji secara klinikal dan beberapa masih sedang dikaji pada biologi leiomioma.

Untuk menimbang perubahan genetik pada leiomioma berbanding miometrium,terapi gene dapat menjanjikan target untuk menghentikan pembentukan leiomioma.Laporan telah menunjukkan terapi dengan adenovirus mediated herpes simplex virus thymidine

kinase/ganciclovir menginhibisi proliferasi petumbuhan leiomioma pada manusia dan tikus dan mengurangkan volume fibroid uteri pada model tikus Eker. Tambahan pula, Ad- mediated delivery of a DNER (dominant negative E receptor) gene juga mengecilkan tumor leiomioma uteri pada tikus Eker. Selain faktor genetik,pemahaman tentang status metilasi DNA dan modifikasi histone pada patogenesis leiomioma uteri adalah kritikal dalam mengembangkan terapi epigenetik yang akan memulihkan modifikasi reguler pola epigenetik dengan menginhibisi enzim epigenetik modifier ( DNA methltransferases, demethylases, and histone deacetylases). Dengan munculnya miRNA sebagai kunci regulator kepada kestabilan ekspesi gene,ini akan lebih menarik untuk diselidiki tentang potensi peran regulasi miRNAs terhadap target gene terpilih.yang dimana produknya dapat mempengaruhi pertumbuhan leiomioma. Dengan mempertimbangkan peran paling penting hormon seks steroid pada pertumbuhan leiomioma, GnRH agonis telah diluluskan oleh US Food and Drug Administration kerana mengurangkan volume fibroid dan simptom yang terkait. US Food and Drug Administration juga telah meluluskan alat intrauterine levenorgestrel-releasing intrauterine system (Mirena) (Bayer Schering Pharma, Germany) sebagai tambahan untuk merawat perdarahan berat pada pengguna alat sahaja.Tambahan lagi beberapa terapi yang mungkin terutama antiprogestin (mifepristone) dan selective Progesterone receptor modulators (asoprisnil, CDB-2914, and CDB4124) telah menunjukkan hasil teraputik yang efesien dan sekarang diuji klinis (www.fda.gov).

Karena sistesis estrogen aromatase dan diekspresi berlebihan pada leiomioma berbanding miometrium pada African American relatif kepada Caucasian American dan wanita jepang beberapa aromatase inhibitors (letrozole, anastrozole, and fadrozole)telah diuji terhadap leiomioma. Sehingga sekarang, trial klinik (fase 1) terhadap letrozole telah ditarik. Namun trial klinikal fase III terhadap anastrozole telah direkrut. Namun beberapa terapi medikal (GnRH antagonists, raloxifene, cabergoline, danazol gestrinone, and lanreotide) telah gagal dalam mencapai efektifitasnya terhadap regresi fiboid dan simptom yang berkatian dengan firboid. Terbukti bahwa growth factor, sitokin dan kemokin dipertimbangkan sebagai efektor potensial estrogen dan progesteron terhadap pertumbuhan leiomioma. Growth factor, sitokin dan kemokin mengawal beberapa proses biologi seperti proliferasi sel,remodelling ECM,angiogenesis dan apoptosis,yang mana merupakan penting dalam pertumbuhan leiomioma. Oleh itu adalah jelas bahwa growth factor, sitokin dan kemokin boleh dipetimbangkan sebagai target major dalam mengawal pertumbuhan leiomioma, Karena EGF telah ditunjukkan dapat memediasi kerja estrogen dan memainkan peran dalam meregulasi pertumbuhan leiomioma. AG1478 dan TKS050 (EGFR blocker) telah ditunjukkan efektif dalam memhentikan pertumbuhan sel leiomioma.TGF-B juga mempunyai peran kritikal terhadap pertumbuhan sel leiomioma. Bukti dari model hewan coba yang rentan secaa genetik juga menunjukkan bahwa Sinyal TGF-B adalah kritikal untuk mempertahankan penyakit ini. Oleh kerna itu telah ditemukan bahwan inhibitor selektif terhadap TGF-RI menurunkan jumlah insidens dan saiz dari tumor ini. Dengan mempertimbangkan bahwa leiomioma dikarekterisasi oleh pertambahan proliferasi sel dan tisu fibrosi, antiproliferasi dan atau agen anti fibrotik boleh menjadi target untuk terapi

leiomioma. Awalnya beberapa campuran

termasuk pirfenidone (235), CP8947 (236),

rosiglitazone (237), ciglitizone (238), halofuginone (239), tranilast semua-trans retinoic acid 242, 243, heparin (244), isoliquiritigenin (245), curcumin 246, 247 dan vitamin D 248, 249, 250,telah menunjukkan efek antiproliferatif dan efek antifibrotik terhadap sel leiomioma. Dari semua ini trial klinik fase 2 terhadap perfenidone telah selesai. Vitamin D menunjukkan hasil yang baik terhadap efektifitas terapi. Vitamin D menginhibisi proliferasi sel leiomioma melalui supresi terhadap COMT. Polimorfisme COMT dan asosiasinya terhadap uterine fibroid adalah lebih tinggi pada African American berbanding wanita berkulit putih atau wanita hispanik Tambah3an lagi defisiensi vitamin D adalah lebih banyak pada African American berbanding European American. Vitamin D juga mengurangkan ekspresi gene TGF3 induced fibrosis related pada sel leiomioma. Tambahan lagi vitamin D mengecilkan saiz tumor pada model tikus Eker. Epigallocatechin gallate yang ditemukan pada teh hijau (Camellia sinensis),secara invitro menunjukkan berlaku apoptosis dan efek inhibisi terhadap proliferasi sel leiomioma.

Eksperimen in vivo menunjukkan bahwa epigallocatechin gallate mengurangkan volume dan berat tumor pada tikus percobaan. Trial klinik rehadap epigallocatechin gallate juga telah selesai. Masih didalam investigasi bahwa leiomioma adalah hasil daripada respon inflamasi yang tidak benar,oleh itu anti-inflamasi (alami atau sintetik) adalah agen efektif untuk tumor jinak ini.

Referensi
Stewart EA. Uterine fibroids. Lancet. 2001;357:293298 View In ArticleAbstractFull Text Full-Text PDF (841 KB) CrossRef Day Baird D, Dunson DB, Hill MC, Cousins D, Schectman JM. High cumulative incidence of uterine leiomyoma in black and white women: ultrasound evidence. Am J Obstet Gynecol. 2003;188:100107 View In ArticleAbstractFull Text Full-Text PDF (324 KB) CrossRef Buttram VC, Reiter RC. Uterine leiomyomata: etiology, symptomatology, and management. Fertil Steril. 1981;36:433445 View In ArticleMEDLINE Marshall LM, Spiegelman D, Barbieri RL, Goldman MB, Manson JE, Colditz GA, et al. Variation in the incidence of uterine leiomyoma among premenopausal women by age and race. Obstet Gynecol. 1997;90:967973 View In ArticleMEDLINECrossRef Wise LA, Palmer JR, Harlow BL, Spiegelman D, Stewart EA, Adams-Campbell LL, et al. Risk of uterine leiomyomata in relation to tobacco, alcohol and caffeine consumption in the Black Womens Health Study. Hum Reprod. 2004;19:17461754 View In ArticleMEDLINECrossRef Walker CL, Stewart EA. Uterine fibroids: the elephant in the room. Science. 2005;308:15891592 View In ArticleCrossRef Flake GP, Andersen J, Dixon D. Etiology and pathogenesis of uterine leiomyomas: a review. Environ Health Perspect. 2003;111:10371054 View In ArticleMEDLINECrossRef Okolo S. Incidence, aetiology and epidemiology of uterine fibroids. Best Pract Res Clin Obstet Gynaecol. 2008;22:571588 View In ArticleAbstractFull Text Full-Text PDF (221 KB) CrossRef Wise LA, Palmer JR, Spiegelman D, Harlow BL, Stewart EA, Adams-Campbell LL, et al. Influence of body size and body fat distribution on risk of uterine leiomyomata in US black women. Epidemiology. 2005;16:346354 View In ArticleMEDLINECrossRef Ye Y, Cheng X, Luo HB, Liu L, Li YB, Hou YP. CYP1A1 and CYP1B1 genetic polymorphisms and uterine leiomyoma risk in Chinese women. J Assist Reprod Genet. 2008;25:389394 View In ArticleCrossRef Baro MA, Oliveira E, Vieira Gomes MT, Cotrim Guerreiro da Silva ID, Ferreira Sartori MG, Batista Castello Giro MJ, et al. The role of MSP I CYP1A1 gene polymorphism in the development of uterine fibroids. Fertil Steril. 2010;94:27832785 View In ArticleAbstractFull Text Full-Text PDF (72 KB) CrossRef Cong RJ, Huang ZY, Cong L, Ye Y, Wang Z, Zha L, et al. Polymorphisms in genes HSD17B1 and HSD17B2 and uterine leiomyoma risk in Chinese women. Arch Gynecol Obstet. 2012;286:701705

View In ArticleCrossRef Ishikawa H, Reierstad S, Demura M, Rademaker AW, Kasai T, Inoue M, et al. High aromatase expression in uterine leiomyoma tissues of African-American women. J Clin Endocrinol Metab. 2009;94:17521756 View In ArticleCrossRef Olmos Grings A, Lora V, Dias Ferreira G, Simoni Brum I, von Eye Corleta H, Capp E. Protein expression of estrogen receptors and and aromatase in myometrium and uteri ne leiomyoma. Gynecol Obstet Invest. 2012;73:113117 View In ArticleCrossRef Al-Hendy A, Salama SA. Ethnic distribution of estrogen receptor- polymorphism is associated with a higher prevalence of uterine leiomyomas in black Americans. Fertil Steril. 2006;86:686693 View In ArticleAbstractFull Text Full-Text PDF (134 KB) CrossRef Al-Hendy A, Salama SA. Catechol-O-methyltransferase polymorphism is associated with increased uterine leiomyoma risk in different ethnic groups. J Soc Gynecol Investig. 2006;13:136144 View In ArticleCrossRef Othman EER, Al-Hendy A. Molecular genetics and racial disparities of uterine leiomyomas. Best Pract Res Clin Obstet Gynaecol. 2008;22:589601 View In ArticleAbstractFull Text Full-Text PDF (292 KB) CrossRef Salama SA, Ho SL, Wang HQ, Tenhunen J, Tilgmann C, Al-Hendy A. Hormonal regulation of catechol-O-methyl transferase activity in women with uterine leiomyomas. Fertil Steril. 2006;86:259262 View In ArticleAbstractFull Text Full-Text PDF (177 KB) CrossRef Gooden KM, Schroeder JC, North KE, Gammon MD, Hartmann KE, Taylor J, et al. Val153Met polymorphism of catechol-O-methyltransferase and prevalence of uterine leiomyomata. Reprod Sci. 2007;14:117120 View In ArticleCrossRef Townsend DE, Sparkes RS, Baluda MC, McClelland G. Unicellular histogenesis of uterine leiomyomas as determined by electrophoresis by glucose-6-phosphate dehydrogenase. Am J Obstet Gynecol. 1970;107:11681173 View In ArticleMEDLINE Mashal RD, Fejzo MLS, Friedman AJ, Mitchner N, Nowak RA, Rein MS, et al. Analysis of androgen receptor DNA reveals the independent clonal origins of uterine leiomyomata and the secondary nature of cytogenetic aberrations in the development of leiomyomata. Genes Chromosomes Cancer. 1994;11:16 View In ArticleMEDLINECrossRef Linder D, Gartler SM. Glucose-6-phosphate dehydrogenase mosaicism: utilization as a cell marker in the study of leiomyomas. Science. 1965;150:6769 View In ArticleMEDLINE Walker CL, Hunter D, Everitt JI. Uterine leiomyoma in the Eker rat: a unique model for important diseases of women. Genes Chromosomes Cancer. 2003;38:349356 View In ArticleMEDLINECrossRef

Cook JD, Walker CL. The Eker rat: establishing a genetic paradigm linking renal cell carcinoma and uterine leiomyoma. Curr Mol Med. 2004;4:813824 View In ArticleMEDLINECrossRef Kiechle-Schwarz M, Sreekantaiah C, Berger CS, Pedron S, Medchill MT, Surti U, et al. Nonrandom cytogenetic changes in leiomyomas of the female genitourinary tract. A report of 35 cases. Cancer Genet Cytogenet. 1991;53:125136 View In ArticleMEDLINECrossRef Rein MS, Friedman AJ, Barbieri RL, Pavelka K, Fletcher JA, Morton CC. Cytogenetic abnormalities in uterine leiomyomata. Obstet Gynecol. 1991;77:923926 View In ArticleMEDLINE Meloni AM, Surti U, Contento AM, Davare J, Sandberg AA. Uterine leiomyomas: cytogenetic and histologic profile. Obstet Gynecol. 1992;80:209217 View In ArticleMEDLINE Sandberg AA. Updates on the cytogenetics and molecular genetics of bone and soft tissue tumors: leiomyoma. Cancer Genet Cytogenet. 2005;158:126 View In ArticleFull Text Full-Text PDF (825 KB) CrossRef Pandis N, Heim S, Bardi G, Floderus UM, Willen H, Mandahl N, et al. Chromosome analysis of 96 uterine leiomyomas. Cancer Genet Cytogenet. 1991;55:1118 View In ArticleMEDLINECrossRef Dal Cin P, Moerman P, Deprest J, Brosens I, Van den Berghe H. A new cytogenetic subgroup in uterine leiomyoma is characterized by a deletion of the long arm of chromosome 3. Genes Chromosomes Cancer. 1995;13:219220 View In ArticleMEDLINECrossRef Nibert M, Heim S. Uterine leiomyoma cytogenetics. Genes Chromosomes Cancer. 1990;2:313 View In ArticleMEDLINECrossRef El-Gharib MN, Elsobky ES. Cytogenetic aberrations and the development of uterine leiomyomata. J Obstet Gynaecol Res. 2010;36:101107 View In ArticleCrossRef Cha PC, Takahashi A, Hosono N, Low SK, Kamatani N, Kubo M, et al. A genome-wide association study identifies three loci associated with susceptibility to uterine fibroids. Nat Genet. 2011;43:447450 View In ArticleCrossRef Gross KL, Neskey DM, Manchanda N, Weremowicz S, Kleinman MS, Nowak RA, et al. HMGA2 expression in uterine leiomyomata and myometrium: quantitative analysis and tissue culture studies. Genes Chromosomes Cancer. 2003;38:6879 View In ArticleMEDLINECrossRef Velagaleti GV, Tonk VS, Hakim NM, Wang X, Zhang H, Erickson-Johnson MR, et al. Fusion of HMGA2 to COG5 in uterine leiomyoma. Cancer Genet Cytogenet. 2011;202:1116 View In ArticleAbstractFull Text Full-Text PDF (647 KB) CrossRef Hunter DS, Klotzbcher M, Kugoh H, Cai SL, Mullen JP, Manfioletti G, et al. Aberrant expression of HMGA2 in uterine leiomyoma associated with loss of TSC2 tumor suppressor gene function. Cancer Res. 2002;62:37663772 View In ArticleMEDLINE

Peng Y, Laser J, Shi G, Mittal K, Melamed J, Lee P, et al. Antiproliferative effects by Let-7 repression of high-mobility group A2 in uterine leiomyoma. Mol Cancer Res. 2008;6:663673 View In ArticleCrossRef Wang T, Zhang X, Obijuru L, Laser J, Aris V, Lee P, et al. A micro-RNA signature associated with race, tumor size, and target gene activity in human uterine leiomyomas. Genes Chromosomes Cancer. 2007;46:336347 View In ArticleMEDLINECrossRef Ingraham SE, Lynch RA, Kathiresan S, Buckler AJ, Menon AG. hREC2, a RAD51-like gene, is disrupted by t(12;14) (q15;q24.1) in a uterine leiomyoma. Cancer Genet Cytogenet. 1999;115:5661 View In ArticleAbstractFull Text Full-Text PDF (539 KB) CrossRef Takahashi T, Nagai N, Oda H, Ohama K, Kamada N, Miyagawa K. Evidence for RAD51L1/HMGIC fusion in the pathogenesis of uterine leiomyoma. Genes Chromosomes Cancer. 2001;30:196 201 View In ArticleMEDLINECrossRef Schoenmakers EF, Huysmans C, Van de Ven WJ. Allelic knockout of novel splice variants of human recombination repair gene RAD51B in t(12;14) uterine leiomyomas. Cancer Res. 1999;59:1923 View In ArticleMEDLINE Nezhad MH, Drieschner N, Helms S, Meyer A, Tadayyon M, Klemke M, et al. 6p21 rearrangements in uterine leiomyomas targeting HMGA1. Cancer Genet Cytogenet. 2010;203:247252 View In ArticleAbstractFull Text Full-Text PDF (567 KB) CrossRef Ligon AH, Scott IC, Takahara K, Greenspan DS, Morton CC. PCOLCE deletion and expression analyses in uterine leiomyomata. Cancer Genet Cytogenet. 2002;137:133137 View In ArticleAbstractFull Text Full-Text PDF (360 KB) CrossRef Ptacek T, Song C, Walker CL, Sell SM. Physical mapping of distinct 7q22 deletions in uterine leiomyoma and analysis of a recently annotated 7q22 candidate gene. Cancer Genet Cytogenet. 2007;174:116120 View In ArticleAbstractFull Text Full-Text PDF (291 KB) CrossRef Mkinen N, Heinonen HR, Moore S, Tomlinson IPM, van der Spuy ZM, Aaltonen LA. MED12 exon 2 mutations are common in uterine leiomyomas from South African patients. Oncotarget. 2011;2:966969 View In Article Mkinen N, Mehine M, Tolvanen J, Kaasinen E, Li Y, Lehtonen HJ, et al. MED12, the mediator complex subunit 12 gene, is mutated at high frequency in uterine leiomyomas. Science. 2011;334:252255 View In ArticleCrossRef Je EM, Kim MR, Min KO, Yoo NJ, Lee SH. Mutational analysis of MED12 exon 2 in uterine leiomyoma and other common tumors. Int J Cancer. 2012;131:E1044E1047 View In ArticleCrossRef Markowski DN, Bartnitzke S, Lning T, Drieschner N, Helmke BM, Bullerdiek J. MED12 mutations in uterine fibroids-their relationship to cytogenetic subgroups. Int J Cancer. 2012;131:1528 1536

View In ArticleCrossRef McGuire MM, Yatsenko A, Hoffner L, Jones M, Surti U, Rajkovic A. Whole exome sequencing in a random sample of North American women with leiomyomas identifies MED12 mutations in majority of uterine leiomyomas. PloS One. 2012;7:e33251 View In ArticleCrossRef Prot G, Croce S, Ribeiro A, Lagarde P, Velasco V, Neuville A, et al. MED12 alterations in both human benign and malignant uterine soft tissue tumors. PloS One. 2012;7:e40015 View In ArticleCrossRef Thompson CM, Koleske AJ, Chao DM, Young RA. A multisubunit complex associated with the RNA polymerase II CTD and TATA-binding protein in yeast. Cell. 1993;73:13611375 View In ArticleMEDLINECrossRef Taatjes DJ. The human Mediator complex: a versatile, genome-wide regulator of transcription. Trends Biochem Sci. 2010;35:315322 View In ArticleCrossRef Egger G, Liang G, Aparicio A, Jones PA. Epigenetics in human disease and prospects for epigenetic therapy. Nature. 2004;429:457463 View In ArticleCrossRef Tost J. DNA methylation: an introduction to the biology and the disease-associated changes of a promising biomarker. Mol Biotechnol. 2010;44:7181 View In ArticleCrossRef Li E, Bestor TH, Jaenisch R. Targeted mutation of the DNA methyltransferase gene results in embryonic lethality. Cell. 1992;69:915926 View In ArticleMEDLINECrossRef Shiota K. DNA methylation profiles of CpG islands for cellular differentiation and development in mammals. Cytogenet Genome Res. 2004;105:325334 View In ArticleCrossRef Shiota K, Yanagimachi R. Epigenetics by DNA methylation for development of normal and cloned animals. Differentiation. 2002;69:162166 View In ArticleMEDLINECrossRef Li S, Chiang T, Richard-Davis G, Barrett JC, McLachlan JA. DNA hypomethylation and imbalanced expression of DNA methyltransferases (DNMT1, 3A, and 3B) in human uterine leiomyoma. Gynecol Oncol. 2003;90:123130 View In ArticleMEDLINECrossRef Asada H, Yamagata Y, Taketani T, Matsuoka A, Tamura H, Hattori N, et al. Potential link between estrogen receptor- gene hypomethylation and uterine fibroid formation. Mol Hum Reprod. 2008;14:539545 View In ArticleCrossRef Yamagata Y, Maekawa R, Asada H, Taketani T, Tamura I, Tamura H, et al. Aberrant DNA methylation status in human uterine leiomyoma. Mol Hum Reprod. 2009;15:259267 View In ArticleCrossRef Maekawa R, Yagi S, Ohgane J, Yamagata Y, Asada H, Tamura I, et al. Disease-dependent differently methylated regions (D-DMRs) of DNA are enriched on the X chromosome in uterine leiomyoma. J Reprod Dev. 2011;57:604612

View In ArticleCrossRef Navarro A, Yin P, Monsivais D, Lin SM, Du P, Wei JJ, et al. Genome-wide DNA methylation indicates silencing of tumor suppressor genes in uterine leiomyoma. PloS One. 2012;7:e33284 View In ArticleCrossRef Kouzarides T. Chromatin modifications and their function. Cell. 2007;128:693705 View In ArticleMEDLINECrossRef Wei LH, Torng PL, Hsiao SM, Jeng YM, Chen MW, Chen CA. Histone deacetylase 6 regulates estrogen receptor in uterine leiomyoma. Reprod Sci. 2011;18:755762 View In ArticleCrossRef Walker CL. Epigenomic reprogramming of the developing reproductive tract and disease susceptibility in adulthood. Birth Defects Res A Clin Mol Teratol. 2011;91:666671 View In ArticleCrossRef Greathouse KL, Bredfeldt T, Everitt JI, Lin K, Berry T, Kannan K, et al. Environmental estrogens differentially engage the histone methyltransferase EZH2 to increase risk of uterine tumorigenesis. Mol Cancer Res. 2012;10:546557 View In ArticleCrossRef Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281297 View In ArticleMEDLINECrossRef Miska EA. How microRNAs control cell division, differentiation and death. Curr Opin Genet Dev. 2005;15:563568 View In ArticleCrossRef Marsh EE, Lin Z, Yin P, Milad M, Chakravarti D, Bulun SE. Differential expression of microRNA species in human uterine leiomyoma versus normal myometrium. Fertil Steril. 2008;89:17711776 View In ArticleAbstractFull Text Full-Text PDF (337 KB) CrossRef Pan Q, Luo X, Chegini N. Differential expression of microRNAs in myometrium and leiomyomas and regulation by ovarian steroids. J Cell Mol Med. 2008;12:227240 View In ArticleCrossRef Georgieva B, Milev I, Minkov I, Dimitrova I, Bradford AP, Baev V. Characterization of the uterine leiomyoma microRNAome by deep sequencing. Genomics. 2012;93:275281 View In Article Chuang TD, Panda H, Luo X, Chegini N. miR-200c is aberrantly expressed in leiomyomas in an ethnic-dependent manner and targets ZEBs, VEGFA, TIMP2, and FBLN5. Endocr Relat Cancer. 2012;19:541556 View In ArticleCrossRef Chuang TD, Luo X, Panda H, Chegini N. miR-93/106b and their host gene, MCM7, are differentially expressed in leiomyomas and functionally target F3 and IL-8. Mol Endocrinol. 2012;26:10281042 View In ArticleCrossRef Pan Q, Luo X, Chegini N. microRNA 21: response to hormonal therapies and regulatory function in leiomyoma, transformed leiomyoma and leiomyosarcoma cells. Mol Hum Reprod. 2010;16:215227

View In ArticleCrossRef Zavadil J, Ye H, Liu Z, Wu JJ, Lee P, Hernando E, et al. Profiling and functional analyses of microRNAs and their target gene products in human uterine leiomyomas. PloS One. 2010;5:e12362 View In ArticleCrossRef Fitzgerald JB, Chennathukuzhi V, Koohestani F, Nowak RA, Christenson LK. Role of microRNA-21 and programmed cell death 4 in the pathogenesis of human uterine leiomyomas. Fertil Steril. 2012;98:726734.e2 View In ArticleAbstractFull Text Full-Text PDF (884 KB) CrossRef Nilsson S, Makela S, Treuter E, Tujague M, Thomsen J, Andersson G, et al. Mechanisms of estrogen action. Physiol Rev. 2001;81:15351565 View In ArticleMEDLINE Andersen J, Barbieri RL. Abnormal gene expression in uterine leiomyomas. J Soc Gynecol Investig. 1995;2:663672 View In ArticleMEDLINECrossRef Maruo T, Ohara N, Wang J, Matsuo H. Sex steroidal regulation of uterine leiomyoma growth and apoptosis. Hum Reprod Update. 2004;10:207220 View In ArticleMEDLINECrossRef Benassayag C, Leroy MJ, Rigourd V, Robert B, Honore JC, Mignot TM, et al. Estrogen receptors (ER/ER) in normal and pathological growth of the human myometrium: pregnancy and leiomyoma. Am J Physiol Endocrinol Metab. 1999;276:E1112E1118 View In Article Kovacs KA, Oszter A, Gocze PM, Kornyei JL, Szabo I. Comparative analysis of cyclin D1 and oestrogen receptor (alpha and beta) levels in human leiomyoma and adjacent myometrium. Mol Hum Reprod. 2001;7:10851091 View In ArticleMEDLINECrossRef Otsuka H, Shinohara M, Kashimura M, Yoshida K, Okamura Y. A comparative study of the estrogen receptor ratio in myometrium and uterine leiomyomas. Int J Gynaecol Obstet. 1989;29:189 194 View In ArticleAbstract Full-Text PDF (1539 KB) CrossRef Sadan O, Van Iddekinge B, Van Gelderen CJ, Savage N, Becker PJ, Van Der Walt LA, et al. Oestrogen and progesterone receptor concentrations in leiomyoma and normal myometrium. Ann Clin Biochem. 1987;24:263267 View In Article Ishikawa H, Ishi K, Serna VA, Kakazu R, Bulun SE, Kurita T. Progesterone is essential for maintenance and growth of uterine leiomyoma. Endocrinology. 2010;151:24332442 View In ArticleCrossRef Gao Z, Matsuo H, Nakago S, Kurachi O, Maruo T. p53 Tumor suppressor protein content in human uterine leiomyomas and its down-regulation by 17 beta-estradiol. J Clin Endocrinol Metab. 2002;87:39153920 View In ArticleCrossRef Gustavsson I, Englund K, Faxen M, Sjoblom P, Lindblom B, Blanck A. Tissue differences but limited sex steroid responsiveness of c-fos and c-jun in human fibroids and myometrium.

Mol Hum Reprod. 2000;6:5559 View In ArticleMEDLINECrossRef Andersen J, Grine E, Eng CL, Zhao K, Barbieri RL, Chumas JC, et al. Expression of connexin-43 in human myometrium and leiomyoma. Am J Obstet Gynecol. 1993;169:12661276 View In ArticleAbstract Full-Text PDF (3605 KB) CrossRef Lee EJ, Bajracharya P, Lee DM, Cho KH, Kim KJ, Bae YK, et al. Gene expression profiles of uterine normal myometrium and leiomyoma and their estrogen responsiveness in vitro. Korean J Pathol. 2010;44:272283 View In Article Swartz CD, Afshari CA, Yu L, Hall KE, Dixon D. Estrogen-induced changes in IGF-I, Myb family and MAP kinase pathway genes in human uterine leiomyoma and normal uterine smooth muscle cell lines. Mol Hum Reprod. 2005;11:441450 View In ArticleMEDLINECrossRef Barbarisi A, Petillo O, Di Lieto A, Melone MA, Margarucci S, Cannas M, et al. 17-beta estradiol elicits an autocrine leiomyoma cell proliferation: evidence for a stimulation of protein kinase-dependent pathway. J Cell Physiol. 2001;186:414424 View In ArticleMEDLINECrossRef Ciarmela P, Bloise E, Gray PC, Carrarelli P, Islam MS, De Pascalis F, et al. Activin-A and myostatin response and steroid regulation in human myometrium: disruption of their signalling in uterine fibroid. J Clin Endocrinol Metab. 2011;96:755765 View In ArticleCrossRef Shimomura Y, Matsuo H, Samoto T, Maruo T. Up-regulation by progesterone of proliferating cell nuclear antigen and epidermal growth factor expression in human uterine leiomyoma. J Clin Endocrinol Metab. 1998;83:21922198 View In ArticleCrossRef Matsuo H, Kurachi O, Shimomura Y, Samoto T, Maruo T. Molecular bases for the actions of ovarian sex steroids in the regulation of proliferation and apoptosis of human uterine leiomyoma. Oncology. 1999;57:4958 View In ArticleCrossRef Nierth-Simpson EN, Martin MM, Chiang TC, Melnik LI, Rhodes LV, Muir SE, et al. Human uterine smooth muscle and leiomyoma cells differ in their rapid 17beta-estradiol signaling: implications for proliferation. Endocrinology. 2009;150:24362445 View In ArticleCrossRef Hermon TL, Moore AB, Yu L, Kissling GE, Castora FJ, Dixon D. Estrogen receptor alpha (ER) phospho-serine-118 is highly expressed in human uterine leiomyomas compared to matched myometrium. Virchows Arch. 2008;453:557569 View In ArticleCrossRef Park SH, Ramachandran S, Kwon SH, Cha SD, Seo EW, Bae I, et al. Upregulation of ATP-sensitive potassium channels for estrogen-mediated cell proliferation in human uterine leiomyoma cells. Gynecol Endocrinol. 2008;24:250256 View In ArticleCrossRef Kastner P, Krust A, Turcotte B, Stropp U, Tora L, Gronemeyer H, et al. Two distinct estrogenregulated promoters generate transcripts encoding the two functionally different human

progesterone receptor forms A and B. EMBO J. 1990;9:16031614 View In ArticleMEDLINE Kim JJ, Sefton EC. The role of progesterone signaling in the pathogenesis of uterine leiomyoma. Mol Cell Endocrinol. 2011;358:223231 View In ArticleCrossRef Brandon DD, Bethea CL, Strawn EY, Novy MJ, Burry KA, Harrington MS, et al. Progesterone receptor messenger ribonucleic acid and protein are overexpressed in human uterine leiomyomas. Am J Obstet Gynecol. 1993;169:7885 View In ArticleAbstract Full-Text PDF (2394 KB) CrossRef Marelli G, Codegoni AM, Bizzi A. Estrogen and progesterone receptors in leiomyomas and normal uterine tissues during reproductive life. Acta Eur Fertil. 1989;20:1922 View In ArticleMEDLINE Viville B, Charnock-Jones DS, Sharkey AM, Wetzka B, Smith SK. Distribution of the A and B forms of the progesterone receptor messenger ribonucleic acid and protein in uterine leiomyomata and adjacent myometrium. Hum Reprod. 1997;12:815822 View In ArticleMEDLINECrossRef Ying Z, Weiyuan Z. Dual actions of progesterone on uterine leiomyoma correlate with the ratio of progesterone receptor A: B. Gynecol Endocrinol. 2009;25:520523 View In ArticleCrossRef Fujimoto J, Hirose R, Ichigo S, Sakaguchi H, Li Y, Tamaya T. Expression of progesterone receptor form A and B mRNAs in uterine leiomyoma. Tumour Biol. 1998;19:126131 View In ArticleMEDLINECrossRef Arici A, Sozen I. Transforming growth factor-beta3 is expressed at high levels in leiomyoma where it stimulates fibronectin expression and cell proliferation. Fertil Steril. 2000;73:1006 1011 View In ArticleAbstractFull Text Full-Text PDF (265 KB) CrossRef Maruo T, Matsuo H, Shimomura Y, Kurachi O, Gao Z, Nakago S, et al. Effects of progesterone on growth factor expression in human uterine leiomyoma. Steroids. 2003;68:817824 View In ArticleCrossRef Yamada T, Nakago S, Kurachi O, Wang J, Takekida S, Matsuo H, et al. Progesterone down-regulates insulin-like growth factor-I expression in cultured human uterine leiomyoma cells. Hum Reprod. 2004;19:815821 View In ArticleMEDLINECrossRef Matsuo H, Maruo T, Samoto T. Increased expression of Bcl-2 protein in human uterine leiomyoma and its up-regulation by progesterone. J Clin Endocrinol Metab. 1997;82:293299 View In ArticleCrossRef Kurachi O, Matsuo H, Samoto T, Maruo T. Tumor necrosis factor- expression in human uterine leiomyoma and its down-regulation by progesterone. J Clin Endocrinol Metab. 2001;86:2275 2280 View In ArticleCrossRef Yin P, Lin Z, Cheng YH, Marsh EE, Utsunomiya H, Ishikawa H, et al. Progesterone receptor regulates Bcl-2 gene expression through direct binding to its promoter region in uterine leiomyoma cells. J Clin Endocrinol Metab. 2007;92:44594466

View In ArticleCrossRef Hoekstra AV, Sefton EC, Berry E, Lu Z, Hardt J, Marsh E, et al. Progestins activate the AKT pathway in leiomyoma cells and promote survival. J Clin Endocrinol Metab. 2009;94:1768 1774 View In ArticleCrossRef Luo X, Yin P, Reierstad S, Ishikawa H, Lin Z, Pavone ME, et al. Progesterone and mifepristone regulate L-type amino acid transporter 2 and 4F2 heavy chain expression in uterine leiomyoma cells. J Clin Endocrinol Metab. 2009;94:45334539 View In ArticleCrossRef Yin P, Lin Z, Reierstad S, Wu J, Ishikawa H, Marsh EE, et al. Transcription factor KLF11 integrates progesterone receptor signaling and proliferation in uterine leiomyoma cells. Cancer Res. 2010;70:17221730 View In ArticleCrossRef Sozen I, Arici A. Interactions of cytokines, growth factors, and the extracellular matrix in the cellular biology of uterine leiomyomata. Fertil Steril. 2002;78:112 View In ArticleAbstractFull Text Full-Text PDF (126 KB) CrossRef Ciarmela P, Wiater E, Vale W. Activin-A in myometrium: characterization of the actions on myometrial cells. Endocrinology. 2008;149:25062516 View In ArticleCrossRef Ciarmela P, Wiater E, Smith SM, Vale W. Presence, actions, and regulation of myostatin in rat uterus and myometrial cells. Endocrinology. 2009;150:906914 View In ArticleCrossRef Ren Y, Yin H, Tian R, Cui L, Zhu Y, Lin W, et al. Different effects of epidermal growth factor on smooth muscle cells derived from human myometrium and from leiomyoma. Fertil Steril. 2011;96:10151020 View In ArticleAbstractFull Text Full-Text PDF (615 KB) CrossRef Rossi MJ, Chegini N, Masterson BJ. Presence of epidermal growth factor, platelet-derived growth factor, and their receptors in human myometrial tissue and smooth muscle cells: their action in smooth muscle cells in vitro. Endocrinology. 1992;130:17161727 View In ArticleMEDLINECrossRef Fayed YM, Tsibris JC, Langenberg PW, Robertson AL. Human uterine leiomyoma cells: binding and growth responses to epidermal growth factor, platelet-derived growth factor, and insulin. Lab Invest. 1989;60:3037 View In ArticleMEDLINE Liang M, Wang H, Zhang Y, Lu S, Wang Z. Expression and functional analysis of platelet-derived growth factor in uterine leiomyomata. Cancer Biol Ther. 2006;5:2833 View In ArticleMEDLINECrossRef Arici A, Sozen I. Expression, menstrual cycle-dependent activation, and bimodal mitogenic effect of transforming growth factor-beta1 in human myometrium and leiomyoma. Am J Obstet Gynecol. 2003;188:7683 View In ArticleAbstractFull Text Full-Text PDF (1031 KB) CrossRef Suo G, Jiang Y, Cowan B, Wang JY. Platelet-derived growth factor C is upregulated in human uterine fibroids and regulates uterine smooth muscle cell growth. Biol Reprod. 2009;81:749

758 View In ArticleCrossRef Mesquita FS, Dyer SN, Heinrich DA, Bulun SE, Marsh EE, Nowak RA. Reactive oxygen species mediate mitogenic growth factor signaling pathways in human leiomyoma smooth muscle cells. Biol Reprod. 2010;82:341351 View In ArticleCrossRef Tang XM, Dou Q, Zhao Y, McLean F, Davis J, Chegini N. The expression of transforming growth factor-beta s and TGF-beta receptor mRNA and protein and the effect of TGF-beta s on human myometrial smooth muscle cells in vitro. Mol Hum Reprod. 1997;3:233240 View In ArticleMEDLINECrossRef Lee BS, Nowak RA. Human leiomyoma smooth muscle cells show increased expression of transforming growth factor-beta 3 (TGF beta 3) and altered responses to the antiproliferative effects of TGF beta. J Clin Endocrinol Metab. 2001;86:913920 View In ArticleCrossRef Battegay EJ, Raines EW, Seifert RA, Bowen-Pope DF, Ross R. TGF-beta induces bimodal proliferation of connective tissue cells via complex control of an autocrine PDGF loop. Cell. 1990;63:515524 View In ArticleMEDLINECrossRef Joseph DS, Malik M, Nurudeen S, Catherino WH. Myometrial cells undergo fibrotic transformation under the influence of transforming growth factor beta-3. Fertil Steril. 2010;93:15001508 View In ArticleAbstractFull Text Full-Text PDF (501 KB) CrossRef Wolanska M, Bankowski E. Transforming growth factor beta and platelet-derived growth factor in human myometrium and in uterine leiomyomas at various stages of tumour growth. Eur J Obstet Gynecol Reprod Biol. 2007;130:238244 View In ArticleAbstractFull Text Full-Text PDF (548 KB) CrossRef Ding L, Xu J, Luo X, Chegini N. Gonadotropin releasing hormone and transforming growth factor beta activate mitogen-activated protein kinase/extracellularly regulated kinase and differentially regulate fibronectin, type I collagen, and plasminogen activator inhibitor-1 expression in leiomyoma and myometrial smooth muscle cells. J Clin Endocrinol Metab. 2004;89:55495557 View In ArticleCrossRef Levens E, Luo X, Ding L, Williams RS, Chegini N. Fibromodulin is expressed in leiomyoma and myometrium and regulated by gonadotropin-releasing hormone analogue therapy and TGF-beta through Smad and MAPK-mediated signalling. Mol Hum Reprod. 2005;11:489 494 View In ArticleMEDLINECrossRef Luo X, Ding L, Chegini N. CCNs, fibulin-1C and S100A4 expression in leiomyoma and myometrium: inverse association with TGF-beta and regulation by TGF-beta in leiomyoma and myometrial smooth muscle cells. Mol Hum Reprod. 2006;12:245256 View In ArticleMEDLINE Ciarmela P, Islam MS, Reis FM, Gray PC, Bloise E, Petraglia F, et al. Growth factors and myometrium: biological effects in uterine fibroid and possible clinical implications. Hum Reprod Update. 2011;17:772790

View In ArticleCrossRef Luo X, Ding L, Xu J, Chegini N. Gene expression profiling of leiomyoma and myometrial smooth muscle cells in response to transforming growth factor-beta. Endocrinology. 2005;146:1097 1118 View In ArticleMEDLINECrossRef Strawn EY, Novy MJ, Burry KA, Bethea CL. Insulin-like growth factor I promotes leiomyoma cell growth in vitro. Am J Obstet Gynecol. 1995;172:18371843discussion 18434 View In ArticleAbstract Full-Text PDF (740 KB) CrossRef Van Der Ven L, Gloudemans T, Roholl PJM, Van Buul-Offers SC, Bladergroen BA, Welters MJP, et al. Growth advantage of human leiomyoma cells compared to normal smooth-muscle cells due to enhanced sensitivity toward insulin-like growth factor I. Int J Cancer. 1994;59:427 434 View In ArticleMEDLINECrossRef Gao Z, Matsuo H, Wang Y, Nakago S, Maruo T. Up-regulation by IGF-I of proliferating cell nuclear antigen and Bcl-2 protein expression in human uterine leiomyoma cells. J Clin Endocrinol Metab. 2001;86:55935599 View In ArticleCrossRef Yu L, Saile K, Swartz CD, He H, Zheng X, Kissling GE, et al. Differential expression of receptor tyrosine kinases (RTKs) and IGF-I pathway activation in human uterine leiomyomas. Mol Med. 2008;14:264275 View In Article Peng L, Wen Y, Han Y, Wei A, Shi G, Mizuguchi M, et al. Expression of insulin-like growth factors (IGFs) and IGF signaling: molecular complexity in uterine leiomyomas. Fertil Steril. 2009;91:26642675 View In ArticleAbstractFull Text Full-Text PDF (696 KB) CrossRef Rauk PN, Surti U, Roberts JM, Michalopoulos G. Mitogenic effect of basic fibroblast growth factor and estradiol on cultured human myometrial and leiomyoma cells. Am J Obstet Gynecol. 1995;173:571577 View In ArticleAbstract Full-Text PDF (708 KB) CrossRef Anania CA, Stewart EA, Quade BJ, Hill JA, Nowak RA. Expression of the fibroblast growth factor receptor in women with leiomyomas and abnormal uterine bleeding. Mol Hum Reprod. 1997;3:685691 View In ArticleMEDLINECrossRef Hong T, Shimada Y, Uchida S, Itami A, Li Z, Ding Y, et al. Expression of angiogenic factors and apoptotic factors in leiomyosarcoma and leiomyoma. Int J Mol Med. 2001;8:141148 View In ArticleMEDLINE Arita S, Kikkawa F, Kajiyama H, Shibata K, Kawai M, Mizuno K, et al. Prognostic importance of vascular endothelial growth factor and its receptors in the uterine sarcoma. Int J Gynecol Cancer. 2005;15:329336 View In ArticleCrossRef Yoshida M, Ohtsuru A, Samejima T, Okazaki M, Fujishita A, Ito M, et al. Involvement of parathyroid hormone-related peptide in cell proliferation activity of human uterine leiomyomas. Endocr J. 1999;46:8190

View In ArticleMEDLINECrossRef Weir EC, Goad DL, Daifotis AG, Burtis WJ, Dreyer BE, Nowak RA. Relative overexpression of the parathyroid hormone-related protein gene in human leiomyomas. J Clin Endocrinol Metab. 1994;78:784789 View In ArticleCrossRef Austin DJ, Nowak RA, Stewart EA. Onapristone suppresses prolactin production in explant cultures of leiomyoma. Gynecol Obstet Invest. 1999;47:268271 View In ArticleMEDLINECrossRef Gellersen B, Bonhoff A, Hunt N, Bohnet HG. Decidual-type prolactin expression by the human myometrium. Endocrinology. 1991;129:158168 View In ArticleMEDLINECrossRef Sozen I, Olive DL, Arici A. Expression and hormonal regulation of monocyte chemotactic protein1 in myometrium and leiomyomata. Fertil Steril. 1998;69:10951102 View In ArticleAbstractFull Text Full-Text PDF (3284 KB) CrossRef Pekonen F, Nyman T, Rutanen EM. Differential expression of mRNAs for endothelin-related proteins in human endometrium, myometrium and leiomyoma. Mol Cell Endocrinol. 1994;103:165170 View In ArticleMEDLINECrossRef Robin P, Chouayekh S, Bole-Feysot C, Leiber D, Tanfin Z. Contribution of phospholipase D in endothelin-1-mediated extracellular signal-regulated kinase activation and proliferation in rat uterine leiomyoma cells. Biol Reprod. 2005;72:6977 View In ArticleMEDLINECrossRef Horiuchi A, Nikaido T, Yoshizawa T, Itoh K, Kobayashi Y, Toki T, et al. HCG promotes proliferation of uterine leiomyomal cells more strongly than that of myometrial smooth muscle cells in vitro. Mol Hum Reprod. 2000;6:523528 View In ArticleMEDLINECrossRef Tsai SJ, Lin SJ, Cheng YM, Chen HM, Wing LYC. Expression and functional analysis of pituitary tumor transforming growth factor-1 in uterine leiomyomas. J Clin Endocrinol Metab. 2005;90:37153723 View In ArticleCrossRef Parkin J, Cohen B. An overview of the immune system. Lancet. 2001;357:17771789 View In ArticleAbstractFull Text Full-Text PDF (1185 KB) CrossRef Dinarello CA. Historical insights into cytokines. Eur J Immunol. 2007;37:S34S45 View In ArticleCrossRef Hatthachote P, Gillespie JI. Complex interactions between sex steroids and cytokines in the human pregnant myometrium: evidence for an autocrine signaling system at term. Endocrinology. 1999;140:25332540 View In ArticleMEDLINECrossRef Litovkin KV, Domenyuk VP, Bubnov VV, Zaporozhan VN. Interleukin-6 -174G/C polymorphism in breast cancer and uterine leiomyoma patients: a population-based case control study. Exp Oncol. 2007;29:295298 View In Article Ding L, Luo X, Chegini N. The expression of IL-13 and IL-15 in leiomyoma and myometrium and

their influence on TGF- and proteases expression in leiomyoma and myometrial smooth muscle cells and SKLM, leiomyosarcoma cell line. J Soc Gynecol Invest. 2004;11:319A View In Article Zheng T, Oh MH, Oh SY, Schroeder JT, Glick AB, Zhu Z. Transgenic expression of interleukin-13 in the skin induces a pruritic dermatitis and skin remodeling. J Invest Dermatol. 2008;129:742751 View In Article Sugimoto R, Enjoji M, Nakamuta M, Ohta S, Kohjima M, Fukushima M, et al. Effect of IL-4 and IL13 on collagen production in cultured LI90 human hepatic stellate cells. Liver Int. 2005;25:420428 View In ArticleMEDLINE Fichtner-Feigl S, Strober W, Kawakami K, Puri RK, Kitani A. IL-13 signaling through the IL-132 receptor is involved in induction of TGF-1 production and fibrosis. Nat Med. 2005;12:99 106 View In ArticleMEDLINECrossRef Chen Q, Rabach L, Noble P, Zheng T, Lee CG, Homer RJ, et al. IL-11 receptor in the pathogenesis of IL-13-induced inflammation and remodeling. J Immunol. 2005;174:23052313 View In ArticleMEDLINE Nair S, Al-Hendy A. Adipocytes enhance the proliferation of human leiomyoma cells via TNF- proinflammatory cytokine. Reprod Sci. 2011;18:11861192 View In ArticleCrossRef Hsieh YY, Chang CC, Tsai FJ, Lin CC, Yeh LS, Tsai CH. Tumor necrosis factor-[alpha]-308 promoter and p53 codon 72 gene polymorphisms in women with leiomyomas. Fertil Steril. 2004;82:11771181 View In ArticleAbstractFull Text Full-Text PDF (81 KB) CrossRef Hsieh YY, Chang CC, Tsai CH, Lin CC, Tsai FJ. Interleukin (IL)-12 receptor 1 codon 378 G homozygote and allele, but not IL-1 (-511 promoter, 3953 exon 5, receptor antagonist), IL2 114, IL-4-590 intron 3, IL-8 3-UTR 2767, and IL-18 105, are associated with higher susceptibility to leiomyoma. Fertil Steril. 2007;87:886895 View In ArticleAbstractFull Text Full-Text PDF (156 KB) CrossRef Pietrowski D, Thewes R, Sator M, Denschlag D, Keck C, Tempfer C. Uterine leiomyoma is associated with a polymorphism in the interleukin 1-beta gene. Am J Reprod Immunol. 2009;62:112117 View In Article Sosna O, Kolesar L, Slavcev A, Skibova J, Fait T, Mara M, et al. Th1/Th2 cytokine gene polymorphisms in patients with uterine fibroid. Folia Biol (Praha). 2010;56:206210 View In Article Xing Z, Gauldie J, Tremblay GM, Hewlett BR, Addison C. Intradermal transgenic expression of granulocyte-macrophage colony-stimulating factor induces neutrophilia, epidermal hyperplasia, Langerhans cell/macrophage accumulation, and dermal fibrosis. Lab Invest. 1997;77:615622 View In ArticleMEDLINE Xing Z, Tremblay GM, Sime PJ, Gauldie J. Overexpression of granulocyte-macrophage colony-

stimulating factor induces pulmonary granulation tissue formation and fibrosis by induction of transforming growth factor-beta 1 and myofibroblast accumulation. Am J Pathol. 1997;150:59 View In ArticleMEDLINE Xing Z, Ohkawara Y, Jordana M, Graham F, Gauldie J. Transfer of granulocyte-macrophage colony-stimulating factor gene to rat lung induces eosinophilia, monocytosis, and fibrotic reactions. J Clin Invest. 1996;97:11021110 View In ArticleMEDLINECrossRef Andreutti D, Gabbiani G, Neuville P. Early granulocyte-macrophage colony-stimulating factor expression by alveolar inflammatory cells during bleomycin-induced rat lung fibrosis. Laboratory investigation. 1998;78:14931502 View In Article Chegini N, Tang XM, Ma C. Regulation of transforming growth factor-beta1 expression by granulocyte macrophage-colony-stimulating factor in leiomyoma and myometrial smooth muscle cells. J Clin Endocrinol Metab. 1999;84:41384143 View In ArticleCrossRef Suzuki M, Takamizawa S, Nomaguchi K, Suzu S, Yamada M, Igarashi T, et al. Erythropoietin synthesis by tumour tissues in a patient with uterine myoma and erythrocytosis. Br J Haematol. 2001;113:4951 View In ArticleMEDLINECrossRef Yokoyama Y, Shinohara A, Hirokawa M, Maeda N. Erythrocytosis due to an erythropoietinproducing large uterine leiomyoma. Gynecol Obstet Invest. 2003;56:179183 View In ArticleMEDLINECrossRef Mehrad B, Keane MP, Strieter RM. Chemokines as mediators of angiogenesis. Thromb Haemost. 2007;97:755762 View In ArticleMEDLINE Broxmeyer HE. Chemokines in hematopoiesis. Curr Opin Hematol. 2008;15:4958 View In ArticleCrossRef Bonfield TL, Panuska JR, Konstan MW, Hilliard KA, Hilliard JB, Ghnaim H, et al. Inflammatory cytokines in cystic fibrosis lungs. Am J Respir Crit Care Med. 1995;152:21112118 View In ArticleCrossRef Senturk LM, Sozen I, Gutierrez L, Arici A. Interleukin 8 production and interleukin 8 receptor expression in human myometrium and leiomyoma. Am J Obstet Gynecol. 2001;184:559566 View In ArticleAbstractFull Text Full-Text PDF (213 KB) CrossRef Syssoev KA, Kulagina NV, Chukhlovin AB, Morozova EB, Totolian AA. Expression of mRNA for chemokines and chemokine receptors in tissues of the myometrium and uterine leiomyoma. Bull Exp Biol Med. 2008;145:8489 View In ArticleCrossRef Stewart EA, Friedman AJ, Peck K, Nowak RA. Relative overexpression of collagen type I and collagen type III messenger ribonucleic acids by uterine leiomyomas during the proliferative phase of the menstrual cycle. J Clin Endocrinol Metab. 1994;79:900906 View In ArticleCrossRef Wolanska M, Sobolewski K, Drodewicz M, Bakowski E. Extracellular matrix components in

uterine leiomyoma and their alteration during the tumour growth. Mol Cell Biochem. 1998;189:145152 View In ArticleMEDLINECrossRef Norian JM, Malik M, Parker CY, Joseph D, Leppert PC, Segars JH, et al. Transforming growth factor beta3 regulates the versican variants in the extracellular matrix-rich uterine leiomyomas. Reprod Sci. 2009;16:11531164 View In ArticleCrossRef Malik M, Norian J, McCarthy-Keith D, Britten J, Catherino WH. Why leiomyomas are called fibroids: the central role of extracellular matrix in symptomatic women. Semin Reprod Med. 2010;28:169179 View In ArticleCrossRef Iwahashi M, Muragaki Y. Increased type I and V collagen expression in uterine leiomyomas during the menstrual cycle. Fertil Steril. 2011;95:21372139 View In ArticleAbstractFull Text Full-Text PDF (147 KB) CrossRef Moore AB, Yu L, Swartz CD, Zheng X, Wang L, Castro L, et al. Human uterine leiomyoma-derived fibroblasts stimulate uterine leiomyoma cell proliferation and collagen type I production, and activate RTKs and TGF beta receptor signaling in coculture. Cell Commun Signal. 2010;8:10 View In Article Leppert PC, Baginski T, Prupas C, Catherino WH, Pletcher S, Segars JH. Comparative ultrastructure of collagen fibrils in uterine leiomyomas and normal myometrium. Fertil Steril. 2004;82(Suppl 3):11821187 View In ArticleAbstractFull Text Full-Text PDF (504 KB) CrossRef Mitropoulou TN, Theocharis AD, Stagiannis KD, Karamanos NK. Identification, quantification and fine structural characterization of glycosaminoglycans from uterine leiomyoma and normal myometrium. Biochimie. 2001;83:529536 View In ArticleMEDLINECrossRef Berto AGA, Sampaio LO, Franco CRC, Cesar RM, Michelacci YM. A comparative analysis of structure and spatial distribution of decorin in human leiomyoma and normal myometrium. Biochim Biophys Acta. 2003;1619:98112 View In ArticleMEDLINE Catherino WH, Leppert PC, Stenmark MH, Payson M, Potlog-Nahari C, Nieman LK, et al. Reduced dermatopontin expression is a molecular link between uterine leiomyomas and keloids. Genes Chromosomes Cancer. 2004;40:204217 View In ArticleMEDLINECrossRef Fujita M. Histological and biochemical studies of collagen in human uterine leiomyomas. Hokkaido Igaku Zasshi. 1985;60:602615 View In ArticleMEDLINE Hulboy DL, Rudolph LA, Matrisian LM. Matrix metalloproteinases as mediators of reproductive function. Mol Hum Reprod. 1997;3:2745 View In ArticleMEDLINECrossRef Chegini N. Proinflammatory and profibrotic mediators: principal effectors of leiomyoma development as a fibrotic disorder. Semin Reprod Med. 2010;28:180203

View In ArticleCrossRef Ohara N. Sex steroidal modulation of collagen metabolism in uterine leiomyomas. Clin Exp Obstet Gynecol. 2009;36:1011 View In Article Gelse K, Pschl E, Aigner T. Collagens-structure, function, and biosynthesis. Adv Drug Deliv Rev. 2003;55:15311546 View In ArticleCrossRef Wang H, Mahadevappa M, Yamamoto K, Wen Y, Chen B, Warrington JA, et al. Distinctive proliferative phase differences in gene expression in human myometrium and leiomyomata. Fertil Steril. 2003;80:266276 View In ArticleFull Text Full-Text PDF (43 KB) CrossRef Iwahashi M, Muragaki Y, Ikoma M, Mabuchi Y, Kobayashi AYA, Tanizaki Y, et al. Immunohistochemical analysis of collagen expression in uterine leiomyomata during the menstrual cycle. Exp Ther Med. 2010;2:287290 View In Article Rogers R, Norian J, Malik M, Christman G, Abu-Asab M, Chen F, et al. Mechanical homeostasis is altered in uterine leiomyoma. Am J Obstet Gynecol. 2008;198:474.e1474.e11 View In ArticleAbstractFull Text Full-Text PDF (1859 KB) Summary PDF (201 KB) CrossRef Norian JM, Owen CM, Taboas J, Korecki C, Tuan R, Malik M, et al. Characterization of tissue biomechanics and mechanical signaling in uterine leiomyoma. Matrix Biol. 2012;31:5765 View In ArticleCrossRef Malik M, Segars J, Catherino WH. Integrin 1 regulates leiomyoma cytoskeletal integrity and growth. Matrix Biol. 2012;31:389397 View In ArticleCrossRef Tsibris J, Segars J, Coppola D, Mane S, Wilbanks GD, OBrien WF, et al. Insights from gene arrays on the development and growth regulation of uterine leiomyomata. Fertil Steril. 2002;78:114121 View In ArticleAbstractFull Text Full-Text PDF (102 KB) CrossRef Arslan AA, Gold LI, Mittal K, Suen TC, Belitskaya-Levy I, Tang MS, et al. Gene expression studies provide clues to the pathogenesis of uterine leiomyoma: new evidence and a systematic review. Hum Reprod. 2005;20:852863 View In ArticleMEDLINECrossRef Ahn WS, Kim KW, Bae SM, Yoon JH, Lee JM, Namkoong SE, et al. Targeted cellular process profiling approach for uterine leiomyoma using cDNA microarray, proteomics and gene ontology analysis. Int J Exp Pathol. 2003;84:267279 View In ArticleMEDLINECrossRef Catherino W, Salama A, Potlog-Nahari C, Leppert P, Tsibris J, Segars J. Gene expression studies in leiomyomata: new directions for research. Semin Reprod Med. 2004;22:8390 View In ArticleMEDLINECrossRef Malik M, Catherino WH. Novel method to characterize primary cultures of leiomyoma and myometrium with the use of confirmatory biomarker gene arrays. Fertil Steril. 2007;87:11661172 View In ArticleAbstractFull Text Full-Text PDF (1374 KB) CrossRef

David AC, Sam M, Nichole MB, William WH, Arnold IC. Proteoglycans of uterine fibroids and keloid scars: similarity in their proteoglycan composition. Biochem J. 2012;443:361368 View In ArticleCrossRef Leppert PC, Catherino WH, Segars JH. A new hypothesis about the origin of uterine fibroids based on gene expression profiling with microarrays. Am J Obstet Gynecol. 2006;195:415420 View In ArticleAbstractFull Text Full-Text PDF (384 KB) CrossRef Berto AGA, Oba SM, Michelacci YM, Sampaio LO. Galactosaminoglycans from normal myometrium and leiomyoma. Braz J Med Biol Res. 2001;34:633637 View In Article Rein MS, Barbieri RL, Welch W, Gleason RE, Caulfield JP, Friedman AJ. The concentrations of collagen-associated amino acids are higher in GnRH agonist-treated uterine myomas. Obstet Gynecol. 1993;82:901905 View In ArticleMEDLINE Hjelm A, Ekman-Ordeberg G, Barchan K, Malmstrm A. Identification of the major proteoglycans from human myometrium. Acta Obstet Gynecol Scand. 2001;80:10841090 View In ArticleMEDLINECrossRef Dou Q, Tarnuzzer RW, Williams RS, Schultz GS, Chegini N. Differential expression of matrix metalloproteinases and their tissue inhibitors in leiomyomata: a mechanism for gonadotrophin releasing hormone agonist-induced tumour regression. Mol Hum Reprod. 1997;3:10051014 View In ArticleMEDLINECrossRef Bodner-Adler B, Bodner K, Kimberger O, Czerwenka K, Leodolter S, Mayerhofer K. Expression of matrix metalloproteinases in patients with uterine smooth muscle tumors: an immunohistochemical analysis of MMP-1 and MMP-2 protein expression in leiomyoma, uterine smooth muscle tumor of uncertain malignant potential, and leiomyosarcoma. J Soc Gynecol Investig. 2004;11:182186 View In ArticleMEDLINECrossRef Wolanska M, Sobolewski K, Bakowski E, Jaworski S. Matrix metalloproteinases of human leiomyoma in various stages of tumor growth. Gynecol Obstet Invest. 2004;58:1418 View In ArticleMEDLINECrossRef Bogusiewicz M, Stryjecka-Zimmer M, Postawski K, Jakimiuk AJ, Rechberger T. Activity of matrix metalloproteinase-2 and-9 and contents of their tissue inhibitors in uterine leiomyoma and corresponding myometrium. Gynecol Endocrinol. 2007;23:541546 View In ArticleCrossRef Maybin JA, Critchley HOD, Jabbour HN. Inflammatory pathways in endometrial disorders. Mol Cell Endocrinol. 2011;335:4251 View In ArticleCrossRef Fujii S, Suzuki A, Matsumura N, Kanamori T, Shime H, Fukuhara K, et al. Fibroids: basic science and etiology. International Congress Series. Amsterdam: Elsevier; 2004; View In Article Kanamori T, Takakura K, Mandai M, Kariya M, Fukuhara K, Kusakari T, et al. PEP-19 overexpression in human uterine leiomyoma. Mol Hum Reprod. 2003;9:709717 View In ArticleMEDLINECrossRef

Fukuhara K, Kariya M, Kita M, Shime H, Kanamori T, Kosaka C, et al. Secreted frizzled related protein 1 is overexpressed in uterine leiomyomas, associated with a high estrogenic environment and unrelated to proliferative activity. J Clin Endocrinol Metab. 2002;87:1729 1736 View In ArticleCrossRef Jabbour HN, Sales KJ, Catalano RD, Norman JE. Inflammatory pathways in female reproductive health and disease. Reproduction. 2009;138:903919 View In Article Eckes B, Colucci-Guyon E, Smola H, Nodder S, Babinet C, Krieg T, et al. Impaired wound healing in embryonic and adult mice lacking vimentin. J Cell Sci. 2000;113:24552462 View In Article Wynn TA. Cellular and molecular mechanisms of fibrosis. J Pathol. 2007;214:199210 View In ArticleCrossRef Desmouliere A, Darby IA, Gabbiani G. Normal and pathologic soft tissue remodeling: role of the myofibroblast, with special emphasis on liver and kidney fibrosis. Lab Invest. 2003;83:1689 1707 View In ArticleMEDLINECrossRef Guarino M, Tosoni A, Nebuloni M. Direct contribution of epithelium to organ fibrosis: epithelialmesenchymal transition. Hum Pathol. 2009;40:13651376 View In ArticleAbstractFull Text Full-Text PDF (2737 KB) CrossRef Schiller M, Javelaud D, Mauviel A. TGF-beta-induced SMAD signaling and gene regulation: consequences for extracellular matrix remodeling and wound healing. J Dermatol Sci. 2004;35:8392 View In ArticleAbstractFull Text Full-Text PDF (318 KB) CrossRef Hinz B. Tissue stiffness, latent TGF-1 Activation, and mechanical signal transduction: implications for the pathogenesis and treatment of fibrosis. Curr Rheumatol Rep. 2009;11:120126 View In Article Ono M, Maruyama T, Masuda H, Kajitani T, Nagashima T, Arase T, et al. Side population in human uterine myometrium displays phenotypic and functional characteristics of myometrial stem cells. Proc Natl Acad Sci U S A. 2007;104:1870018705 View In ArticleCrossRef Chang HL, Senaratne TN, Zhang LH, Szotek PP, Stewart E, Dombkowski D, et al. Uterine leiomyomas exhibit fewer stem/progenitor cell characteristics when compared with corresponding normal myometrium. Reprod Sci. 2010;17:158167 View In ArticleCrossRef Ono M, Qiang W, Serna VA, Yin P, Coon JS, Navarro A, et al. Role of stem cells in human uterine leiomyoma growth. PloS one. 2012;7:e36935 View In Article Mas A, Cervell I, Gil-Sanchis C, Faus A, Ferro J, Pellicer A, et al. Identification and characterization of the human leiomyoma side population as putative tumor-initiating cells. Fertil Steril. 2012;98:741751.e6 View In ArticleAbstractFull Text Full-Text PDF (1787 KB) CrossRef

Feinberg EC, Larsen FW, Catherino WH, Zhang J, Armstrong AY. Comparison of assisted reproductive technology utilization and outcomes between Caucasian and African American patients in an equal-access-to-care setting. Fertil Steril. 2006;85:888894 View In ArticleAbstractFull Text Full-Text PDF (111 KB) CrossRef Ezzati M, Norian JM, Segars JH. Management of uterine fibroids in the patient pursuing assisted reproductive technologies. Womens Health. 2009;5:413421 View In Article Levy G, Hill MJ, Beall S, Zarek SM, Segars JH, Catherino WH. Leiomyoma: genetics, assisted reproduction, pregnancy and therapeutic advances. J Assist Reprod Genet. 2012;29:703712 View In ArticleCrossRef Salama SA, Kamel M, Christman G, Wang HQ, Fouad HM, Al-Hendy A. Gene therapy of uterine leiomyoma: adenovirus-mediated herpes simplex virus thymidine kinase/ganciclovir treatment inhibits growth of human and rat leiomyoma cells in vitro and in a nude mouse model. Gynecol Obstet Invest. 2007;63:6170 View In ArticleMEDLINECrossRef Hassan M, Zhang D, Salama S, Hamada F, Arafa H, Fouad H, et al. Towards fibroid gene therapy: adenovirus-mediated delivery of herpes simplex virus 1 thymidine kinase gene/ganciclovir shrinks uterine leiomyoma in the Eker rat model. Gynecol Obstet Invest. 2009;68:1932 View In ArticleCrossRef Hassan MH, Salama SA, Zhang D, Arafa HMM, Hamada F, Fouad H, et al. Gene therapy targeting leiomyoma: adenovirus-mediated delivery of dominant-negative estrogen receptor gene shrinks uterine tumors in Eker rat model. Fertil Steril. 2010;93:239250 View In ArticleAbstractFull Text Full-Text PDF (1238 KB) CrossRef Shushan A, Rojansky N, Laufer N, Klein BY, Shlomai Z, Levitzki R, et al. The AG1478 tyrosine kinase inhibitor is an effective suppressor of leiomyoma cell growth. Hum Reprod. 2004;19:19571967 View In ArticleMEDLINECrossRef Shushan A, Ben-Bassat H, Mishani E, Laufer N, Klein BY, Rojansky N. Inhibition of leiomyoma cell proliferation in vitro by genistein and the protein tyrosine kinase inhibitor TKS050. Fertil Steril. 2007;87:127135 View In ArticleAbstractFull Text Full-Text PDF (900 KB) CrossRef Laping NJ, Everitt JI, Frazier KS, Burgert M, Portis MJ, Cadacio C, et al. Tumor-specific efficacy of transforming growth factor-beta RI inhibition in Eker rats. Clin Cancer Res. 2007;13:3087 3099 View In ArticleMEDLINECrossRef Lee BS, Margolin SB, Nowak RA. Pirfenidone: a novel pharmacological agent that inhibits leiomyoma cell proliferation and collagen production. J Clin Endocrinol Metab. 1998;83:219 223 View In ArticleCrossRef Catherino WH, Malik M, Driggers P, Chappel S, Segars J, Davis J. Novel, orally active selective progesterone receptor modulator CP8947 inhibits leiomyoma cell proliferation without adversely affecting endometrium or myometrium. J Steroid Biochem Mol Biol. 2010;122:279 286

View In ArticleCrossRef Zhang C, Wen Z, Li J, Li C, Shi M, Yang G, et al. Inhibition of proliferation and transforming growth factor ss3 protein expression by peroxisome proliferators-activated receptor gamma ligands in human uterine leiomyoma cells. Chin Med J (Engl). 2008;121:166171 View In Article Kim BY, Cho CH, Song DK, Mun KC, Suh SI, Kim SP, et al. Ciglitizone inhibits cell proliferation in human uterine leiomyoma via activation of store-operated Ca2+ channels. Am J Physiol Cell Physiol. 2005;288:C389C395 View In ArticleMEDLINECrossRef Grudzien MM, Low PS, Manning PC, Arredondo M, Belton RJ, Nowak RA. The antifibrotic drug halofuginone inhibits proliferation and collagen production by human leiomyoma and myometrial smooth muscle cells. Fertil Steril. 2010;93:12901298 View In ArticleAbstractFull Text Full-Text PDF (810 KB) CrossRef Shime H, Kariya M, Orii A, Momma C, Kanamori T, Fukuhara K, et al. Tranilast inhibits the proliferation of uterine leiomyoma cells in vitro through G1 arrest associated with the induction of p21waf1 and p53. J Clin Endocrinol Metab. 2002;87:56105617 View In ArticleCrossRef Islam MS, Protic O, Stortoni P, Giannubilo S, Ciavattini A, Lamanna P, et al. Antiproliferative effect of tranilast on human myometrial and leiomyoma cells. Biol Biomed Rep. 2012;2:321327 View In Article Boettger-Tong HL, Stancel GM. Retinoic acid inhibits estrogen-induced uterine stromal and myometrial cell proliferation. Endocrinology. 1995;136:29752983 View In ArticleMEDLINECrossRef Malik M, Webb J, Catherino WH. Retinoic acid treatment of human leiomyoma cells transformed the cell phenotype to one strongly resembling myometrial cells. Clin Endocrinol (Oxf). 2008;69:462470 View In ArticleCrossRef Mason HR, Nowak RA, Morton CC, Castellot JJ. Heparin inhibits the motility and proliferation of human myometrial and leiomyoma smooth muscle cells. Am J Pathol. 2003;162:18951904 View In ArticleAbstractFull Text Full-Text PDF (819 KB) CrossRef Kim D, Ramachandran S, Baek S, Kwon SH, Kwon KY, Cha SD, et al. Induction of growth inhibition and apoptosis in human uterine leiomyoma cells by isoliquiritigenin. Reprod Sci. 2008;15:552558 View In ArticleCrossRef Malik M, Britten J, Driggers P, Payson M, Segars JH, Catherino WH. Curcumin, a nutritional supplement with antineoplastic activity, selectively inhibits leiomyoma growth. Fertil Steril. 2007;88:S217 View In ArticleFull Text Full-Text PDF (44 KB) CrossRef Tsuiji K, Takeda T, Li B, Wakabayashi A, Kondo A, Kimura T, et al. Inhibitory effect of curcumin on uterine leiomyoma cell proliferation. Gynecol Endocrinol. 2011;27:512517 View In ArticleCrossRef Bluer M, Rovio PH, Ylikomi T, Heinonen PK. Vitamin D inhibits myometrial and leiomyoma cell proliferation in vitro. Fertil Steril. 2009;91:19191925

View In ArticleAbstractFull Text Full-Text PDF (311 KB) CrossRef Sharan C, Halder SK, Thota C, Jaleel T, Nair S, Al-Hendy A. Vitamin D inhibits proliferation of human uterine leiomyoma cells via catechol-O-methyltransferase. Fertil Steril. 2011;95:247 253 View In ArticleAbstractFull Text Full-Text PDF (661 KB) CrossRef Halder SK, Sharan C, Al-Hendy A. 1,25-dihydroxyvitamin D3 treatment shrinks uterine leiomyoma tumors in the Eker rat model. Biol Reprod. 2012;86:116 View In ArticleCrossRef Nesby-ODell S, Scanlon KS, Cogswell ME, Gillespie C, Hollis BW, Looker AC, et al. Hypovitaminosis D prevalence and determinants among African American and white women of reproductive age: third National Health and Nutrition Examination Survey, 19881994. Am J Clin Nutr. 2002;76:187192 View In ArticleMEDLINE Halder SK, Goodwin JS, Al-Hendy A. 1,25-Dihydroxyvitamin D reduces TGF-3-induced fibrosisrelated gene expression in human uterine leiomyoma cells. J Clin Endocrinol Metab. 2011;96:E754E762 View In ArticleCrossRef Zhang D, Al-Hendy M, Richard-Davis G, Montgomery-Rice V, Rajaratnam V, Al-Hendy A. Antiproliferative and proapoptotic effects of epigallocatechin gallate on human leiomyoma cells. Fertil Steril. 2010;94:18871893 View In ArticleAbstractFull Text Full-Text PDF (1007 KB) CrossRef Zhang D, Al-Hendy M, Richard-Davis G, Montgomery-Rice V, Sharan C, Rajaratnam V, et al. Green tea extract inhibits proliferation of uterine leiomyoma cells in vitro and in nude mice. Am J Obstet Gynecol. 2010;202:289.e1289.e9

Anda mungkin juga menyukai